You are on page 1of 71

cover

29/8/05

5:21 PM

Page 1

An infection during pregnancy that could


damage the fetus or infant is much less
common than suspicion or fear of infection.
Vague, nonspecific symptoms malaise,
aches and pains, headache, tiredness, nausea
- are the hallmarks of many vertically
transmissible maternal infections but they are
also common during pregnancy or in any
busy, stressful life. In pregnant women they
cant be dismissed as trivial as they may be in
others.
This publication represents a consensus
among specialists in perinatal infection. It has
been endorsed by the Australasian Society
for Infectious Diseases and the Royal College
of Obstetricians and Gynaecologists of
Australia and New Zealand. We hope it will
not discourage nonspecialists from referring
pregnant women with suspected infection
during pregnancy, but that it will help to guide
the initial assessment and investigation of the
patient, and prevent unnecessary anxiety or
hasty decisions.

Management of
Perinatal Infections
Cytomegalovirus
Enterovirus
Hepatitis B
Hepatitis C
Herpes Simplex Virus
Human Immunodeficiency Virus
Listeria
Mycobacterium Tuberculosis
Parvovirus
Rubella
Streptococcus - Group B
Toxoplasma
Treponema Pallidum (Syphilis)
Varicella Zoster Virus

Edited by Dr Pamela Palasanthiran,


Dr Mike Starr, and Dr Cheryl Jones
Introduction by Prof Lyn Gilbert
AUSTRALASIAN SOCIETY FOR
INFECTIOUS DISEASES 2002

AUSTRALASIAN SOCIETY FOR


INFECTIOUS DISEASES 2002

Management of
Perinatal Infections
Cytomegalovirus
Enterovirus
Hepatitis B
Hepatitis C
Herpes Simplex Virus
Human Immunodeficiency Virus
Listeria
Mycobacterium Tuberculosis
Parvovirus
Rubella
Streptococcus - Group B
Toxoplasma
Treponema Pallidum (Syphilis)
Varicella Zoster Virus

Edited by Dr Pamela Palasanthiran, Dr


Mike Starr, and Dr Cheryl Jones

AUSTRALASIAN SOCIETY FOR


INFECTIOUS DISEASES 2002

Copyright 2002 All rights reserved.


This book is copyright. Except as permitted under the Copyright Act 1968, (for example a
fair dealing for the purposes of study, research, criticism or review) no part of this book
may be reproduced, stored in a retrieval system, or transmitted in any form or by any
means without prior written permission. All inquiries should be made to Book House at the
address below.
ISBN 1 74018 222 7
Published by the Australasian Society for Infectious Diseases (ASID)
145 Macquarie Street, Sydney NSW 2000
Ph: (02) 9256 5475 Fax: (02) 9256 9692 e-mail: asid@racp.edu.au
www.racp.edu.au/asid
Reprinted in Australia 2005 by Wild & Woolley,
a division of Books & Writers Network Pty Ltd
Watsons Bay
2006:
Some updates and/or emendations have been included for selected algorithms under the
heading "Emendations". The comments are not comprehensive and are meant as added
information pending a new edition of this publication.
The Editors

C O N T E N T S

Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .iv
Foreword . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .v
Dr Pamela Palasanthiran, Dr Mike Starr, Dr Cheryl Jones

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .vii
Prof Lyn Gilbert

Cytomegalovirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Dr Pamela Palasanthiran, Dr Cheryl Jones, Dr Sue Garland

Enterovirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Dr Alison Kesson

Hepatitis B Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .9
Dr Jim Buttery

Hepatitis C Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Dr Jim Buttery

Herpes simplex Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16


Dr Cheryl Jones

Human immunodeficiency Virus . . . . . . . . . . . . . . . . . . . . . . . . . 19


Dr Pamela Palasanthiran

Listeria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Dr Lesley Voss

Mycobacterium tuberculosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Dr Mike Starr & Dr Allen Yung

Parvovirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
Prof Lyn Gilbert & Dr Mike Starr

Rubella . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
Dr Clare Nourse

Streptococcus, group B . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
A/Prof Suzanne M. Garland & Dr Mike Starr

Toxoplasma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
Prof Lyn Gilbert

Treponema pallidum (Syphilis) . . . . . . . . . . . . . . . . . . . . . . . . . .42


Dr Andrew Daley & Prof Lyn Gilbert

Varicella zoster Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45


Dr Anne Marie Heuchan & Prof David Isaacs

References and Acknowledgements . . . . . . . . . . . . . . . . . . . . . . 51

iii

C O N T R I B U T O R S

Dr Jim Buttery, Paediatric Infectious Diseases, Physician, Royal Childrens


Hospital, Melbourne, Vic
Dr Andrew Daley, Clinical Microbiologist, Department of Microbiology and
Infectious Diseases, Womens and Childrens Health, Parkville, Vic
A/Prof Suzanne M. Garland, Director, Department of Microbiology & Infectious
Diseases, Womens and Childrens Health, Parkville, Vic
Prof Lyn Gilbert, Head, Centre for Infectious Diseases & Microbiology, Institute
of Clinical Pathology and Medical Research, Westmead Hospital, Sydney,
NSW
Dr Anne Marie Heuchan, Specialist Registrar in Neonatology, The Royal
Hospital for Sick Children, Glasgow, UK
Prof David Isaacs, Senior Staff Specialist, Department of Immunulogy and
Infectious Diseases, The Childrens Hospital at Westmead, NSW
Dr Cheryl Jones, Paediatric Infectious Diseases Consultant, The Childrens
Hospital at Westmead, NSW
Dr Alison Kesson, Medical Virologist, The Childrens Hospital at Westmead, NSW
Dr Clare Nourse, Paediatric Infectious Diseases Physician, Womens and
Childrens Health Service, Mater Hospital, Brisbane, Qld
Dr Pamela Palasanthiran, Staff Specialist, Paedriatric Infectious Diseases, Sydney
Childrens Hospital, Randwick, NSW
Dr Mike Starr, Paediatric Infectious Diseases Physician, Royal Childrens
Hospital, Melbourne, Vic
Dr Lesley Voss, Paediatric Infectious Diseases Physician, Starship Children's
Hospital, Auckland, NZ
Dr Allen Yung, Infectious Diseases Physician, Victorian Infectious Diseases
Service, Melbourne, Vic

iv

F O R E W O R D

This comprehensive set of contemporary algorithms on the


management of perinatal infections has been written by a team of
Australian and New Zealand paediatric infectious disease experts who
are all members of the Australasian Society of Infectious Diseases
(ASID).
The impetus behind this project was the recognition that simple
guidelines are needed for general practitioners, obstetricians and
paediatricians to address common issues once an infectious agent
complicates a pregnancy. Questions such as Do I really have this
infection? Can it be treated during my pregnancy? Will I pass it on to
my baby? and What will happen if I do? frequently arise. Thus, the
algorithms follow 4 themes (where possible): antenatal diagnosis of
the infectious agent, antenatal management, perinatal transmission
risks, intervention strategies if available, and management of the
newborn. It is stressed that these algorithms are evidence-based
where possible but in many cases, represent consensus
recommendations and are intended only as a set of guidelines.
We acknowledge the contributions of Drs. Sonia Grover and Ted
Weaver who reviewed the final versions of these algorithms and
provided invaluable feedback on behalf of the Royal Australian and
New Zealand College of Obstetricians and Gynaecologists
(RANZCOG) and members of ASID who provided comments and
feedback on early drafts. We also wish to thank ASID and in
particular the ASID council for funding for this project.
Above all, we acknowledge the authors for the invaluable time they
have contributed to this project, on top of their primary work
commitments. We also thank Professor Lyn Gilbert for her
encouragement and unfailing guidance. Their combined support has
been a significant force behind this achievement
We hope these algorithms will be as useful to you as they have been
for us.
Pamela Palasanthiran
Mike Starr
Cheryl Jones
March 2002

I N T R O D U C T I O N

An infection during pregnancy that could damage the fetus or infant is


much less common than suspicion or fear of infection. Vague,
nonspecific symptoms malaise, aches and pains, headache,
tiredness, nausea are the hallmarks of many vertically transmissible
maternal infections but they are also common during pregnancy or in
any busy, stressful life. In pregnant women they cant be dismissed as
trivial as they may be in others. Laboratory tests may provide the
answer but can be hard to interpret. On the other hand, vertically
transmissible infections are often asymptomatic. Routine antenatal
screening is recommended for some on the basis of criteria including:
Incidence of maternal infection, transmission to the fetus and
damage if infection occurs.
Availability of a reliable screening test
Availability of a safe effective intervention to prevent fetal/infant
infection and reduce damage.
Sometimes asymptomatic women are screened for infections other
than those recommended. An unexpectedly positive result then
creates a problem that must be managed. It can be difficult to
determine the predictive value of a positive test, in the absence of
symptoms. The result may not indicate current or recent infection, but
this may become apparent only after additional serological testing
and/or examination of amniotic fluid. Meanwhile, it will have caused
anxiety to the patient and her family. Her medical adviser will be
worried, not only for the wellbeing of the patient and her fetus, but also
about the potential for litigation if things go wrong. If the results remain
uncertain as they often do the patient may choose to terminate her
pregnancy because she cannot tolerate even a small risk of having an
abnormal infant. On the other hand, even an inappropriate screening
test will sometimes detect a significant infection and the potential to
intervene and prevent an unfortunate outcome.
Negotiating this minefield of psychological, medicolegal, ethical and
medical dilemmas is difficult enough for the specialist with a particular
interest in infections in pregnancy. But for the average GP or
obstetrician, for whom they are rare complications, it can be nervewracking. These algorithms outline recommended antenatal screening
tests, how to manage symptomatic infection or a positive screening
test and the range of tests available to assist in risk assessment.

vii

There are a few common themes:


Significant decisions about management of a suspected infection
during pregnancy should never be based on the result of single
test. The test should always be repeated, preferably by a different
method and on another specimen of serum. False positive IgM
results are not uncommon and, even in the best laboratories, sera
occasionally get mixed up.
Laboratories are advised to keep serum, sent for routine antenatal
screening, for at least 12 months. If the possibility of infection arises
later it can be retrieved and tested in parallel with a later specimen,
to show or exclude seroconversion, which is much better evidence
of recent infection than IgM.
If only a single serum is available or the results do not change in
serial samples, measuring IgG avidity (a measure of maturity of the
IgG response, which develops over a period of several months)
can help. A high avidity can exclude recent infection, even in the
presence of persisting IgM. The reverse is not necessarily true low avidity does not always imply recent infection.
This publication represents a consensus among specialists in perinatal infection. Its completion is a major achievement for the editors,
Pamela Palasanthiran, Mike Starr and Cheryl Jones. It has been
endorsed by the Australasian Society for Infectious Diseases and the
Royal College of Obstetricians and Gynaecologists of Australia and
New Zealand. We hope it will not discourage nonspecialists from
referring pregnant women with suspected infection during pregnancy,
but that it will help to guide the initial assessment and investigation of
the patient, and prevent unnecessary anxiety or hasty decisions.
Lyn Gilbert
Centre for Infectious Diseases and
Microbiology
Institute of Clinical Pathology and
Medical Research
Westmead Hospital
March 2002

viii

C Y T O M E G A L O V I R U S
A L G O R I T H M

Diagnosis of Suspected Maternal CMV Infection


Possible indications for antenatal testing
Routine antenatal testing?a
History suggestive of CMV illnessb
Exposure to known CMV infected
individual or blood product
Immunocompromised
Abnormalities on routine antenatal
ultrasound (SEE ALGORITHM 2)

LABORATORY
INVESTIGATIONS1

DIRECT VIRUS DETECTION

SEROLOGYc

IgG ve
IgM ve

IgG +ve
IgM ve

NOT CMV INFECTION


but susceptibled

PAST
INFECTION

IgG ve or +ve
IgM +vec

CMV direct
flourescent
antibody
(DFA)

No change
in IgG

Possible
recent
infection

CMV specific
IgG avidityc

Low probable
recent infection

High probable
past infection

RECENT PRIMARY INFECTION


NOTES
a As CMV is the leading cause of congenital
infections
(0.3 2% of live births), antenatal testing
remains a consideration, especially in
counselling issues for primary maternal
CMV infections. If done, there should be
appropriate management plans.
All pregnant women should be advised
about simple infection control
precautions to reduce transmission risk
eg handwashing after nappy changes
and contact with respiratory tract
secretions, especially with children
<2 years of age attending day care
Some groups identified at higher risk of
primary CMV as determined by annual

Nucleic Acid
Detection
PCR (DNA),
NASBA (RNA)

COMMENTS: Relevant samples include buffy coat (white


cells), nasopharyngeal aspirate or saliva and urine
1. Sensitivity for detecting infection:
DFA = 8090%, while PCR = 95100%
2. Sensitivity for detecting disease:
DFA = 7080% compared with PCR = 5090%
3. Specificity for disease: DFA = 95-100%,
virus isolation = 95100% and PCR = 4050%
4. Time to results: DFA and PCR: "rapid";
Virus isolation could take up to 4 weeks

Repeat test 2-4 weeks later

Seroconversion
or rise in IgG

CMV isolation

SEE ALGORITHM 2

+ve

seroconversion rates
specificity). More sensitive assays can
I. Day care workers 11% per annum
detect IgM in 5% for up to 2 years
II. Parents with child in day care 2030%
IgM can persist for months after primary
per annum
infection or reappear with reactivation or
In comparison, healthcare workers
re-infection
seroconvert at a rate comparable to the
False positive IgM occur with crossgeneral population ie 23% per annum
reactivity with other herpes viruses,
b Most primary CMV infections are
pregnancy or autoimmune disorders
asymptomatic. Suspect primary CMV
CMV IgG avidity assay is reported to be
disease in a viral illness associated with
a reliable indicator of primary infection.
atypical lymphocytosis which is "Monospot"
Low avidity = probable recent infection,
negative (also seen in primary
with progression to high avidity with
toxoplasmosis) or with clinical syndromes
time2
associated with CMV disease
d If still concerned or if serology taken within
c Overall, IgM is ~75% sensitive by ELISA
2 weeks of clinical illness, repeat in 2
Commercially available assays: IgM
weeks
results may lack specificity (~75%

C Y T O M E G A L O V I R U S
A L G O R I T H M

Management of
Primary Maternal CMV Infection
CONFIRMED PRIMARY MATERNAL CMV

Fetal Diagnosis

Fetal Risk Assessment


SEE ALGORITHM 3

Therapy
Supportive
None available for either
prevention of transmission or
for effective treatment of
congenital CMV

Serial Fetal
Ultrasoundsa

Invasive in utero
Investigationsb

AMNIOCENTESIS

PCR

FETAL BLOOD SAMPLING

Culture

FBE (Plt)

LFT

Serology
(IgM)

PCR

<20 w

>20 w

<20 w

>20 w

>20 w (Not performed under 21 weeks)

45%

80100%

18%

56%

5080%
Sensitivity of Test

NOTES
a. Features associated with symptomatic congenital CMV infection include:
hydrocephalus (ventricular dilation)
microcephaly
ascites
intrauterine growth retardation (IUGR)
hydrops fetalis
pleural or pericardial effusions
oligo or polyhydramnios intracranial calcification
hepatomegaly
abdominal calcification
pseudomeconium ileus
Caution advised in interpretation of findings as presence of signs not
always predictive of degree of fetal damage. The sensitivity of fetal ultrasound is hard to evaluate from the literature. Overall, ? 3050% sensitive.
b. Invasive in utero investigations35
Techniques available but all lack sensitivity. Experience with fetal blood
sampling is limited.
Diagnosis is best achieved by a combination of fetal ultrasound +

amniocentesis (for PCR) +/- fetal serology


Diagnosis by amniocentesis testing (PCR & culture)
and fetal IgM is about 45% sensitive overall if taken <
20 weeks and 80100% sensitive if taken > 20 weeks
gestation. Specificity approaches 100%
Sensitivity is increased by waiting 6 weeks after
maternal infection
Positive results do NOT predict any degree of fetal
damage
Quantitative PCR and/or culture may identify infected
fetuses at risk of symptomatic disease (direct relationship between high load and risk)6
Further data required before active recommendation.
Positive results: pregnancy termination is an option
by informed choice.

C Y T O M E G A L O V I R U S
A L G O R I T H M

Congenital CMV:
Fetal Risk Assessment
PRIMARYa

NON-PRIMARY
(reinfection or reactivation)

50% risk of transmissionb

1% risk of transmission

Symptomatic
congenital CMV 10%

Risk of
sequelae
90%d

Normal
10%

Asymptomatic
congenital CMV 90%

Risk of
sequelae
10%e

Symptomatic
congenital CMV 1%c

Asymptomatic
congenital CMV 99%

RISK OF SEQUELAE up to 10% e

Normal
90%

Risk of fetal morbidity overall is 2025% SEE ALGORITHM 4

NOTES
a. Australian epidemiology: The incidence of primary CMV infection in pregnancy in Australia is estimated to be 6/1000 pregnancies.
The incidence of congenital CMV is estimated to be 0.3 to 0.6%7
b. The risk of transmission is distributed equally between the 3 trimesters. However,
Risk of severe adverse neurological outcome more likely in 1 infection in first half of pregnancy. Features of fetus affected in early
infancy include small for gestational age (SGA), microcephaly and intracranial calcifications
A fetus affected late in pregnancy is more likely to have acute visceral disease (hepatitis, pneumonia, purpura and severe
thrombocytopenia)
c. Infection of CMV-seropositive women with a different CMV strain can lead to intrauterine transmission and symptomatic congenital
infection.8
d. Main concerns of symptomatic congenital CMV infection are:
1. A mortality rate of between 1030%
2. Neurological sequelae of microcephaly (3550%), seizures (10%), chorioretinitis (1020%), mental retardation (70%)
3. Sensory neural hearing loss (SNHL, 2550%)9
30% )
Unilateral
Bilateral
70% ) Progressive
57% (over 2 to 6 years?)
43% )
Stable
e. Main concerns of asymptomatic congenital CMV are
1.
Sensory neural hearing loss (5%)
Unilateral
64% )
Bilateral
36% ) Progressive
36% (over 2 to 6 years?)
64% )
Stable
If hearing is preserved at age 12 months, intellectual development is unlikely to be affected10
2. Chorioretinitis (2%)11

Emendation 2006
Algorithm 3: Congenital CMV: Fetal risk assessment
Prior maternal CMV immunity (i.e. seropositivity) results in an approximate
70% reduction in the risk of congenital CMV infection.E1 However, it has now
been demonstrated that the risk of congenital CMV infection after primary
maternal CMV infection remains elevated for up to four years postseroconversion, with the highest risk being in the first two years post
seroconversion. The overall risk is 12.7% post seroconversion, that decreases
to the baseline 1% risk by about 4 years post seroconversion.E2 The time of
seroconversion is usually unknown, however, this information may be helpful
when counselling a women of a child with congenital CMV infection about
the time of a subsequent pregnancy

C Y T O M E G A L O V I R U S
A L G O R I T H M

Management of the
Newborn at Risk of Congenital CMV
Thorough physical examination at birtha

Ophthalmology

Radiological
Examination

Head ultrasound
hydrocephalus
(insensitive
investigation
for intracranial
calcification)

+ve

Laboratory Investigations
Must be done 3 weeks of birth

CT of brain
(+/-MRI)
intracranial
calcification
ventriculomegaly
cerebral atrophy

Serology
CMV IgM

ve

+ve

+ve

PCR
buffy coat
(white cells)

Viral culture
Urine
+/- NPA

ve

ve

+ve

+ve

Congenital CMV

SYMPTOMATIC
Intensity of management dependent
on extent of organ involvement
NOTES
a. Features of (typical) symptomatic congenital CMV
1. Neurological: (see text box, Algorithm 3)
2. Non-neurological: SGA (50%), petechiae/purpura
(5075%), hepatosplenomegaly (4060%), neonatal jaundice (4065%), pneumonia (<1%), dental
abnormalities (30%), inguinal hernias (males),
infant mortality (1030%).
3. Laboratory abnormalities: hemolytic anemia (1050%), platelets 75,000/mm (50%), direct hyperbilirubinemia 3 mg/dL (3570%), AST > 100 IU/L
(2580%), CSF protein >120 mg/dL (4550%)
4. Risk of serious bacterial infection (5%)

ASYMPTOMATIC
36 monthly review for first 2 years
including neurodevelopmental assessment

NOTES
1. Other baseline tests at birth: FBE & differential and LFT
2. Hearing tests: due to improved prognosis for speech development if hearing
abnormality detected early, suggest testing before 6 months, then annually
for the first 2 years of life, then close review till age 6 years if no problems
detected
3. All CMV-infected infants should have fundoscopy for retinal lesions and visual assessment by ophthalmologist at diagnosis. If abnormalities detected,
annual review recommended. Otherwise, monitor vision during infancy with
formal review if abnormalities detected.
4. Treatment options: nil. Ganciclovir (IV) for retarding the progress of hearing
loss in CNS disease associated with congenital CMV has been studied, but
follow-up data yet to confirm efficacy12
5. Congenitally infected babies are high CMV shedders. Pregnant health care
workers should be aware of this and infection control practices e.g., handwashing emphasised.

Emendation 2006
Algorithm 4: Management of the newborn
A randomised, controlled trial on intravenous ganciclovir therapy (for 6
weeks) in newborns with symptomatic congenital CMV and CNS disease
showed normal or improved hearing at 6 months (results did not reach
significance) and no hearing deterioration at 1 year (highly significant).
However, the high loss to follow-up (~ 60%) makes the clinical significance of
the data difficult to interpret and 63% had significant neutropenia. Therefore,
the potential side effects and social cost of 6 weeks of intravenous ganciclovir
therapy in an individual child must be weighed against a possible, but yet
unproven long term gain with this treatment.E3 There is currently no indication
for systemic antiviral therapy for infants without clinical signs of CMV
infection at birth (i.e. asymptomatic infection).

E N T E R O V I R U S
A L G O R I T H M

Antenatal Diagnosis of
Maternal Enteroviral Infections
If concerns raised about whether pregnant
woman has an enteroviral infection, are there
features consistent with enterovirus infection?

No

Yes

Observe mother and infant after birth

If there are clinical concerns, the diagnosis can


be made via serology, viral culture (nasopharynx
or throat swab & faeces) and/or PCR

SYMPTOMS OF ENTEROVIRAL DISEASE INCLUDE*:

DIAGNOSIS

Asymptomatic

Serology very insensitive, very specific


Viral culture varying sensitivity and specificity
PCR variable sensitivity

Fever
GIT symptoms eg diarrhoea
Encephalitis

INTERPRETATION OF RESULTS

Flaccid paralysis

Serology:
If the results are positive indicates infection;
if the results are negative infection is not
excluded due to poor sensitivity.

Herpangina
Hand-foot and mouth syndrome
Lymphonodular pharyngitis
Exanthem

Viral Culture:
Sensitivity varies depending on the sample.
Culture has high specificity but due to
prolonged shedding in faeces a viral culture
may be a "false positive" and not indicate a
current enteroviral infection.

Epidemic conjunctivitis
Myositis
Guillain-Barre syndrome
Mononucleosis-like syndrome
Pleurodynia
Acute haemorrhagic conjunctivitis

PCR:
Sensitivity and specificity are good.

Myocardidits

* peak incidence in the Spring/Summer months in non-tropical or temperate regions of Australia

NOTES
Enteroviral infections generally cause insignificant
illness, and perinatal transmission of enteroviruses
leading to significant symptomatic disease in infants is
rare. The literature on this is not comprehensive. Hence,
these algorithms are based largely on anecdotal
evidence and represent broad guidelines.
To date, there are no data on indicators of perinatal
transmission risks nor predictors of fetal or infant
damage.

E N T E R O V I R U S
A L G O R I T H M

Management of
Proven Maternal Infection
Pregnant or postnatal woman has
symptoms and signs consistent with
enteroviral infection which may be
supported by laboratory data

Woman has
mild disease

Woman has
severe disease

Symptomatic
therapy

Consider therapy
with pleconaril

Intrauterine, perinatal and


post-natal transmission occur
No reliable information
about correlates of risk of perinatal
transmission nor predictors
of fetal/infant damage

E N T E R O V I R U S
A L G O R I T H M

Antenatal Management
and Prevention Strategies
Management of
infected mother
Symptomatic
treatment of mother
Counselling

Prevention strategies possible


either before or during
pregnancy, labour or at birth
Polio vaccination prevents
polio. Good personal hygiene
to prevent faecal-oral
transmission especially if in
contact with a source case.
Enteric precautions apply if
patients are admitted.

Interpret clinical and


pathological Information
Risk assessment of fetal
Infection or abnormality:
there are no reliable risk data

Management of
antenatal care
The following has been reported:
abortion, congenital abnormalities,
stillbirth, symptomatic infant,
normal infant

E N T E R O V I R U S
A L G O R I T H M

Diagnosis, Management and Follow-up


of At-Risk Infants
Symptoms and signs of an infected infant
Asymptomatic, Febrile illness,
Sepsis-like illness, Respiratory illness
Herpangina, Coryza, Pharyngitis,
Laryngo-tracheo-bronchitis and bronchitis
Pneumonia, Vomiting and diarrhoea,
Hepatitis, Pancreatitis, Myocarditis
Examthem, Meningitis and encephalitis, SIDS

Diagnosis
Serology, viral culture and PCR

Treatment
Supportive care, polio vaccine,
IVIG may be of benefit if <6 weeks preterm
but no supporting data
Pleconaril limited data available5,6
Counsel mother

Prophylaxis

NOTES
Improved survival in small case
series of severe neonatal Ev
sepsis. Data group was too
small to determine if outcome
was significant. No activity
against Ev 71.5
Recommended neonatal dose
is 5mg/kg given orally every 8
hours. Bioavailability in
neonates is satisfactory.6

Nursery epidemics have been controlled


by infection control using enteric
precautions and prophylactic IVIG.

Follow-up
As clinically indicated by nature
and severity of infection

Emendation 2006
Enterovirus- Algorithm 4: A double blind placebo-controlled trial of oral
Pleconaril in 21 infants with enteroviral meningitis demonstrated plasma levels
sufficient for in vitro inhibition of enterovirus replication.E4 However, oral
Pleconoril has since been withdrawn from the market.

H E P A T I T I S

A L G O R I T H M

V I R U S
1

Antenatal Diagnosis of Hepatitis B


Routine screening recommended
Hepatitis B surface Antigen (HBsAg)

All HepBsAg positive


women require medical
referral post delivery
by midwife/doctor
managing pregnancy

HBsAg +ve

HBsAg ve

Confirm HBsAg+
Further serology
HBeAg
anti-HBeAb
LFTs

Not infected
Nil further action unless
high risk groupa
(offer mother vaccination
after delivery)

HBeAg+ve
High risk carrier

Ensure screening
+/- vaccination of
family members

HBeAg ve

LFTs abnormal

LFTs normal

Consider HBV DNA hybridisation


to detect pre-core mutants. (Available in reference
laboratories only. Turnaround time is slow.)

NOTES
a. High risk groups1
from area of high prevalence
household contact HB carrier
multiple sexual partners
IV drug use
tattoos/body piercing

DNA not
detected

DNA
detected

Low risk
carrier

High risk
carrier

H E P A T I T I S

A L G O R I T H M

V I R U S
2

Management of Hepatitis B in Pregnancy


Acute Hepatitis B

Chronic Hepatitis B infection (HepBsAg+)

SEE ALGORITHM 1

Trimester 1 and 2
perinatal transmission risk ~10%2

Late Pregnancy
perinatal transmission risk ~75% 2

Refer for supportive medical management


No demonstrated role for antenatal HBIG in
reducing subsequent perinatal transmission
Lamivudine (3TC) and -IFN
contra-indicated in pregnancy

Delivery:
no data regarding mode of delivery in
acute hepatitis
Caesarean section lowers risk of perinatal
transmission in chronically infected
HepBeAg positive mothers3

Baby:
HBIG and HB vaccine at birth
further HB vaccine at 2,4 & 12 months
(multivalent vaccines after first dose)5
arrange follow-up serology at 12 months
(SEE ALGORITHM 4)

NOTE
Current prospective data does not support an
increased risk of vertical transmission of Hep B after
amniocentesis.4

10

H E P A T I T I S

V I R U S

A L G O R I T H M

Hepatitis B: Exposure During Pregnancy


No Immunisation

Prior Immunisation

Check serology urgently

Check serology urgently

antiHBs > 10 mIU/mL

HBsAg neg

HBsAg pos

Nil
further
action

Check
HBeAg
status
SEE
ALGORITHM
4

HB vaccine to baby at
0,2,4 & 12 months if
ongoing risk exposure
(multivalent vaccines
after 1st dose)5

antiHBs < 10 mIU/mL

antiHBs > 10 mIU/mL

antiHBs < 10 mIU/mL

HB vaccine
HBIG within 72h if
high risk exposure

Nil further action

HB vaccine at 12
and 6 months5
HBIG within 72h if
high risk exposure

HB vaccine at 12
and 6 months5
Follow-up mother in
3 months: HepBsAg

Follow-up mother in
3 months:
HepBsAg HB vaccine
to baby at birth

HB vaccine to baby at
0,2,4 & 12 months if
ongoing risk exposure
(multivalent vaccines
after 1st dose)5

NOTES
Maternal risk of infection by mode of exposure:
needlestick: if donor eAg+ high risk 20-40%6
sexual contact
mucosal exposure

11

H E P A T I T I S

A L G O R I T H M

V I R U S
4

Management of Infants
of Mothers with Hepatitis B
Maternal serology:
HBsAg positive or
HBeAg positive or
HBV PCR positive

Delivery:
HepBeAg+ mothers: there is no evidence that Caesarean
section offers an additional advantage over the recommended
neonatal reg imen of Ig + vaccine in preventing ver tical
transmissions. A benefit was seen in one study where vaccine
only was used3

HBIG 0.5 mL IM
HB vaccine 0.5 mL IM

At birth: HBIG and HB


vaccine preferably within 12
hours5

Risk vertical transmission:


mother sAg+, eAg- 520%7,8
mother sAg+, eAg+ 7090% 9
90% infected infants become
chronic carriers6

At 2, 4 and 12 months:
HB vaccine
(multivalent schedule)

Breastfeeding:
HBV DNA and HBsAg
detected in breast milk10
no added transmission risk
demonstrated11

Follow-up serology at
12 months including HBsAg

HBsAg negative

HBsAg positive

No
further action

LFTs, HBeAg
Refer for ongoing
management

12

Emendation 2006
HEPATITIS B

Management of a mother exposed to HB during pregnancy (see Algorithm 3).


If maternal antiHBs titre < 10mIU/ml, give mother HBIG (400 IU, IM) as
soon as possible but within 72 hours of exposure. Also, give the mother HB
vaccine within 7 days of exposure and at 1 and 6 months post initial dose.E5

Management of an infant born to a HB carrier mother (see Algorithms 2 & 4):


e.g. mother with acute HBV in pregnancy or is HB antigen or PCR +ve. Give
HBIG (100 IU, IM) to the infant preferably within 12 hours of delivery
(efficacy markedly reduced if administration delayed beyond 48 hours after
birth). Monovalent HB vaccine should be given at the same time (other limb)
if possible, but do not delay beyond 7 days of life. Complete schedule with 3
more doses at 2, 4 and 6 or 12 months (timing dependent on combination
vaccine used). E5

Revised recommendation for primary HB vaccine schedule for babies


(Algorithm 3): when the risk of perinatal HBV transmission is low, the routine
Australian schedule is recommended i.e. birth (monovalent HB vaccine), then
combination vaccines at 2, 4 and 6 or 12 months (timing dependent on
combination vaccine used). E5

H E P A T I T I S

A L G O R I T H M

V I R U S
1

Antenatal Diagnosis of Hepatitis C


Known Hep C Ab +ve

All positive Hep C ELISA


tests need confirmation by
RIBA (recombinant
immunoblot assay) assay
on same sample

Confirm Hep C Ab +ve

All HIV +ve mothers need


Hep C RNA PCR irrespective
of Ab status as serology can
be falsely ve

Status unknown: Indications


for antenatal testing2
IV drug user (past or present)
known abnormal LFTs
previous blood products
before 1990
previous organ transplant or
haemodialysis
partner Hep C positive*
history of incarceration

Counsel re testing for:


Hepatitis B & HIV
Notifiable disease (coded)

Pre-test counselling

Hep C Ab +ve

Counselling
LFTs
Hep C RNA PCR

Hep C RNA PCR ve

Hep C RNA PCR +ve

Low risk carrier1

Demonstrated risk of
vertical transmission1

Refer for medical management


See ALGORITHM 2
Recommend postnatal Hepatitis A vaccination

13

Hep C Ab ve

*partner Hep C positive less


clear risk factor3
nucleotide sequence
homology seen in partners
negligible transmission in
sex-partner studies
Hep C transmission lower
than other sexually
transmitted diseases in high
risk groups

H E P A T I T I S

A L G O R I T H M

V I R U S
2

Management of Hepatitis C in Pregnancy


Hep C Ab +ve

Hep C RNA PCR


LFTs
Refer for management and counselling

HIV and Hepatitis B testing


after counselling

Hep C RNA PCR -ve

Hep C RNA PCR +ve

HIV/Hep C co-infection

Perinatal transmission risk


~0%4

Perinatal transmission risk ~6%4


Risk proportional to RNA load4

Perinatal transmission risk


Hep C 9-45%4

Delivery: no clear evidence re


mode of delivery and reduction
of perinatal transmisison

Breastfeeding: no increased risk


of transmission demonstrated

Interferon-alpha and
ribavirin therapy contraindicated in pregnancy

See ALGORITHM 3

14

H E P A T I T I S

V I R U S

A L G O R I T H M

Management and Follow-up of


Infants of Hepatitis C Infected Mothers

Birth
ensure hepatitis B immunisation within 24 hrs
check maternal records re HBV and HIV testing

Breastfeeding: not discouraged


transmission has not been documented5
Hep C RNA has been detected in breast milk 6
consider expressing and discarding milk if nipples
cracked or bleeding5
consider avoiding breast feeding if symptomatic
liver disease7

Follow-up:
anti Hep C Ab at 1218 months*
or if concerned earlier
anti Hep C Ab
Hep C RNA PCR

Anti Hep C Ab +ve


Infected child*

Anti Hep C Ab ve

Not infected

Hep C RNA PCR


LFTs

Hep C RNA PCR ve

Hep C RNA PCR +ve

Repeat serology
in 3 months

Refer for ongoing


management to
paediatric hepatologist:
repeat LFTs and HCV
RNA PCR
possible therapy

NOTES
Most uninfected infants are antibody negative by 12
months. If positive serology at 12 months, retest in 3
months before considering them infected. A small
percentage of perinatally infected children may clear
the virus1,4,8

15

Emendation 2006
HEPATITIS C
Algorithm 3 : Follow-up of infants of hepatitis C infected mothers E6
The general recommendation for testing a well child with perinatal HCV
exposure is to test the child for HCV antibodies at 18 months of age as
transplacental maternal HCV antibodies should clear by then.
When follow up cannot be guaranteed however, testing by HCV RNA PCR
(include LFT) should be performed earlier, but not at less that one month of age
as the sensitivity of HCV RNA PCR is 22 % at < 1 month of age. A single
positive PCR result after 1 month of age gives a post-test probability of infection
of 73% for a child born to an HIVve/HCV+ve woman, and 90% for a child
born to an HIV+ve/HCV+ve woman.E7 A positive or negative PCR result should
be confirmed on a separate occasion.E8 Negative PCR results but positive anti
HCV antibody in a child < 18 months of age usually suggests that the child is
not infected.8 However, HCV antibody (with accompanying liver function tests)
should be retested at or beyond 18 months of age to confirm this, as occasionally
it represents infection of the child in the absence of HCV viremia. E6, E 8

H E R P E S

S I M P L E X
A L G O R I T H M

V I R U S

Herpes Simplex Virus Infections in Pregnancy:


Risk Assessment of Neonatal Disease
YES

Maternal history of prior


genital HSV infection?

Symptomatic
prior infection

Asymptomatic
prior infection

Genital HSV (1 or 2) diagnosed during pregnancy


HSV culture type & serology determined

Recurrent
infection
(HSV Ab +ve
= HSV type from
genital culture)

Delivered
through a birth
canal with overt
vesicles (Only
20% are culture
positive)1

Overall risk
of neonatal
HSV disease

1%3

NO

No
prior infection

First genital HSV (1 or 2) diagnosed during pregnancy


HSV culture type & serology determined

New
acquisition
of other
HSV type in
genital area

"Initial nonprimary" infection


(HSV Ab -ve to
type cultured
from genital area
& HSV Ab +ve to
other type)

"Primary"
infection (HSV Ab
-ve to both types
+ HSV genital
culture +ve)

Seroconversion well before delivery


(ie prior to 3034 weeks)4

Delivered through a
birth canal with normal
appearance: (Risk of
shedding 1.4%.1 If shedding,
risk of transmission
is 3%, if not it is 0.02%2)

0.023%2

YES (Risk of
shedding 7%)4

NO
(or unknown)

3%4

30-50%1,4

Effect of Caesarean delivery on neonatal attack rate unknown3


NOTES
Most genital HSV infections (primary, non-primary or recurrent) are asymptomatic. ie most mothers of infants with neonatal HSV disease
were previously unaware of their own infection.
HSV-1 genital infection is being increasingly recognised- but is less likely to recur than genital HSV-2.
85% of neonatal HSV infections are acquired perinatally. True intrauterine infection accounts for 5% of reported cases, usually to
women with newly acquired infection. Spontaneous abortion, IUGR, preterm labour have also been reported. These complications are
rare (<1%) for women with primary or recurrent disease.4

16

H E R P E S

S I M P L E X
A L G O R I T H M

V I R U S

Maternal Management of Genital Herpes


Simplex Virus Infections in Pregnancy
History of genital HSV
(laboratory confirmed)

No prior history
of genital HSV

Serial genital cultures not predictive


of shedding during labour,
so are not recommended4

Consider use of suppressive


aciclovir (400 mg tds po) in women
with multiple recurrent overt lesions

First genital HSV infection


diagnosed during pregnancy

Recurrent
infection
(HSV Ab
+ve to type
from genital
culture

First genital
HSV infection
diagnosed
during labour

Obtain HSV serology (type


specific) + type genital culture

New infection (HSV Ab ve


to type from genital culture

In labour:
careful speculum examination

No
active
lesions seen

Proceed
to vaginal
delivery
Fetal scalp
electrode,
forceps,
& vacuum
delivery not
recomended

Management of
newborn as per
ALGORITHM 3

Active
lesions seen
(see notes)

ROM
>6
hours5

ROM
<6
hours5

Diagnosis made early


in pregnancy (1st or
2nd trimester) Counsel
as for ALGORITHM 1

Diagnosis made late


in pregnancy
(ie 3rd trimester)

Seroconversion well
before delivery (ie prior
to 30-34 weeks)4

Consider
suppresssive aciclovir
(400 mg po tds)6

Yes

No or
unknown

Deliver by Caesarean section


Perform genital "sweep culture." If vaginal delivery unavoidable:
fetal scalp electrode, forceps, & vacuum delivery not recommended

NOTES
Insufficient data are available on the effect of suppressive aciclovir on transmission to the newborn for either active recurrent or primary
genital maternal disease.
Careful speculum examination for active genital HSV should be performed on all women at delivery.
ROM>6 hours has been observed in one small study to increase risk of neonatal infection.5 However the efficacy of Caesarean delivery
in preventing neonatal HSV infections is unknown, and this practice has been recently challenged in two large studies in which infected
infants were born in the presence of intact membranes.3

17

Emendation 2006:
HERPES SIMPLEX VIRUS
Algorithm 1 and 2: HSV in pregnancy: risk assessment/management of
neonatal disease
Caesarean section reduces risk of HSV transmission in women shedding HSV
at the time of birth, particularly in women with first time infections who are
HSV type specific antibody negative. E9

H E R P E S

S I M P L E X
A L G O R I T H M

V I R U S

Herpes Simplex Virus Infections


in Pregnancy: Neonatal Management
Low risk
of neonatal HSV disease
ie mother with recurrent
infection, or primary infection
seroconverted well prior
to delivery

Collect surface swabs at 24


hours of life eye, throat,
umbilicus, rectum, urine

High risk of neonatal HSV disease


ie mother with primary infection
close to delivery or infant born
through birth canal with active
HSV disease to mother with no
prior history of genital HSV

Perform:

Follow for signs of infection

LFTs

HSV PCR on blood (if available)

Positive surface swab


or sick newborn

Commence IV aciclovir
immediately, duration
will depend on surface
culture & CSF results

surface swabs at birth


(onset of clinical signs) eye,
throat, umbilicus, rectum, urine
blood count (for low platelets),

Perform lumbar puncture


(CSF analysis, viral culture,
PCR for HSV DNA), blood count
(for low platelets), LFTs, & HSV
PCR on blood (if available)

Infant with
clinical signs of
neonatal HSV disease
(regardless of
maternal history)

Commence IV aciclovir
immediately from birth
If clinical signs of HSV disease
develop, perform LP (CSF
analysis, viral culture, HSC PCR),
CNS imaging and repeat blood
count, LFTs.

Vesicular skin lesions or


atypical pustular or
bullous lesions, especially
on presenting part
Seizures
Unexplained sepsis with
ve blood cultures not
responding to antibiotics

Low platelets

Elevated LFTs

DIC

Respiratory distress
(after day 1 of life)
Corneal ulcer/keratitis

ACICLOVIR THERAPY
Oral therapy should not be used for empiric or
therapeutic treatment of HSV in the neonate.
The current recommended dose is 20 mg/kg/dose IV,
3 times/day as 12 hour infusion. For disease confined to skin,
eye, mouth: duration of therapy is 14 days. For encephalitis,
disseminated disease: 21 days is recommended.7

Follow up:

NOTES
The incidence of neonatal HSV disease in Australia is approximately
3.0 per 100,000 live births.9

Survivors should be monitored closely for recurrences, eye disease,


CNS sequelae. LP should always be performed on all infants with
suspected HSV relapse to exclude CNS involvement.
A phase I/II trial of oral aciclovir to prevent adverse neurological outcome
in infants with frequent cutaneous recurrences after HSV-2 skin, eye,
mouth disease demonstrated neutropenia in 50%, development of
aciclovir-resistant virus in 4% and reduction of cutaneous recurrences
(CNS outcome was not studied).8 Until results of larger, ongoing phase III
trial available, suppressive therapy cannot be routinely recommended.

18

H U M A N

I M M U N O D E F I C I E N C Y
A L G O R I T H M

V I R U S

Diagnosis of HIV Infection


in Pregnant Women
Possible indications for testing
1. Recipient of blood product or human tissue pre1985
2. Past history of intravenous drug use (IDU)
3. Partner of person with IDU or resident/partner from high prevalence area
4. Bisexual relationship
5. Seroconversion illness

Routine antenatal
screening? 1(a)

PRE-TEST COUNSELLING

HIV ANTIBODY
Screen with ELISA
Confirm with Western blot (WB)

ve

+ve

POST-TEST COUNSELLING

Refer to physician specialising


in HIV infection or physician
with access to HIV expertise
Obstetric care in conjunction
with above
Paediatric HIV prenatal counselling
(SEE ALGORITHMS 24)

Lab testing(b)

Should include

HIV RNA
viral load

Repeat in 4 weeks if recent exposure


or indeterminate Western blot

+ve

No further follow up with


respect to HIV unless
re-exposure occurs or risk
behaviour continues

ve

Antiretroviral (ARV) therapy3


see General Principles of ARV in Pregnancy,
ALGORITHM 3

Routine
antenatal
ultrasound
As per routine
obstetric care
unless complications
anticipated

CD4 +ve
lymphocyte subsets

Others:
e.g. FBE, LFT, U&E/Creat

NOTES
a. As effective strategies to prevent perinatal HIV transmission exist, knowing the HIV status in pregnant women may be warranted. Half of HIV +ve pregnant women are unaware of their HIV status
and two-thirds have no risk factors.2
b. Consideration should be given to resistance testing and/or viral subtyping if HIV acquired outside of
Australia.

19

Serology for:
Syphilis
Hep B & C
CMV, HSV
Toxoplasma
Chlamydia screen

H U M A N

I M M U N O D E F I C I E N C Y
A L G O R I T H M

V I R U S

Assessment of Risk Factors in


Perinatal Transmission of HIV
Fetal Risk Assessment & Counselling
Based on figures before the use of routine Highly Active Antiretroviral Therapy (HAART) in management of HIV in adults.

Viral Load
(VL)(HIV RNA
level)

Undetectable
(UD) ie < 400
copies HIV
RNA per mL
by non-ultra
sensitive
assays

Graded levels of
risk, with UD to
1000 copies/mL
carrying the
lowest risk and
> 100,000
copies/mL the
highest risk.
NB: no "safe"
viral load
identified4,5

CD4+
lymphocyte
count

Detectable

Gestation

<37 w

Rupture Of
Membrane (ROM)
> 4 hours

Mode
of delivery

37 w

Breast

Vaginal
(VD)

Approximate
linear inverse
relationship
between CD4
count and risk6

4-fold
in risk7

Approximate
linear inverse
relationship
between
gestational
age and risk6

Mode
of feeding

Caesarean

Doubling of
risk with VD8

Doubling of
risk with breastfeeding9,10

COMMENTS
Other relative risk factors include the first born twin and the presence of co-infections (eg sexually transmitted diseases and
chorioamnionitis).
Antenatal tests as per routine obstetric follow-up. Also, see ALGORITHM 1.
Apart from risk assessment, counselling should include
strategies to prevent transmission (see ALGORITHM 3)
management of baby at birth, including ARV and prophylaxis
testing of baby
care plan before delivery
care plan for future with respect to a family that has HIV "infected" and HIV "affected" members.

20

Bottle

H U M A N

I M M U N O D E F I C I E N C Y
A L G O R I T H M

V I R U S

Strategies to Minimise
the Perinatal Transmission (PNT) of HIV
HIV +ve pregnant women
See General Principles of
ARV in Pregnancy

On antiretroviral therapy (ARV)


(either treatment or prophylaxis)

NO

YES

Presents in labour

Mode of
delivery (a)

Obstetric
care and delivery11

NO

(c)

YES

Formula
feed
IV AZT
Vaginal

VS

+/-

NEVIRAPINE
(NVP)12 (d)

Elective
Caesarean section(b)
IV AZT 3 hours before section
(see Appendix 1)

Caesarean section
Consider if VL >1000
copies/mL or unknown or if
ROM <4 hours (e)

ALGORITHM 4

Follow up with 6
weeks AZT to
infant +/ one
dose NVP if
indicated
ALGORITHM 4

Formula feed
COMMENTS
See Table 1 for estimated PNT risks by selected strategic options.
a If a pregnant woman is already on HAART, the added benefit of caesarean section may be marginal particularly if viral load is undetectable. The combined risk estimates for vertical transmission for women already on an AZT containing regimen is 1 12% (mean
5.7%) with HIV RNA level near delivery of 1000 10, 000 copies/ml and 9 29% (mean 12.6%) if HIV RNA levels are > 10, 000
copies/ml. Based on this, the American College of Obstetricians recommend elective caesarean section as an added strategy if VL >
1000 copies/ml. http://hivatis.org/guidelines
b If labour commences before planned caesarean section and membranes are ruptured for < 4hours, semi-urgent elective caesarean section should be considered (especially if not on ARV prophylaxis and viral load detectable). (see comment e)
c Includes avoidance of invasive procedures e.g. fetal scalp electrodes, episiotomy. If resuscitation needed, gentle oral suction of baby
only (if possible). Wash baby down as soon as possible after birth.
d HIVNET 012: Single dose NVP, 200 mg, to mother intrapartum, and follow up with single dose to infant (2mg/kg) within 3 days of birth.
Caution: Preliminary data report NVP resistance in some mothers and infected infants after a single dose in HIV NET 012 study.
Significance not currently known.14 The role of additional NVP prophylaxis when a woman is already on an established ARV regimen is
unknown and currently not recommended.15
e The value of caesarean section in reducing HIV PNT after a short duration of ROM has not been established. However as a continuum
of PNT risk following ROM exists, some would perform caesarean section to shorten labour and hence duration of ROM.

21

H U M A N

I M M U N O D E F I C I E N C Y
A L G O R I T H M

V I R U S

Management of Infant
at Risk of Perinatal HIV
At Birth Clinical Assessment

a,b

Commence antiretroviral (ARV) prophylaxisc

LABORATORY FOLLOW-UP
See Table 2

PCR
(DNA)

Virus
isolation

CLINICAL

HIV
antibody

Review at times of testing


Clinical examination +
neurodevelopmental assessment
Routine childhood vaccination
except inactivated polio (s/c)
instead of oral polio vaccine
(rationale: HIV infected
household member(s) at risk of
polio from polio faecal excretion
in vaccinee)
Seroreverters: Follow till
adulthood to monitor continued
health after in-utero exposure to
ARV

MEDICATIONS

ARV to stop at 6 weeks


Start co-trimoxazole
(0.5 mLs/kg, daily)
Stop co-trimoxazole when
testing at 3 months confirms
absence of HIV infection in a
non-breast fed infant

COMMENTS
a No HIV embroyapathy syndrome has been described.
b A third of perinatal HIV transmission occurs in utero and 2/3 during the peripartum period. Hence, infected infants are less likely to present
with signs and symptoms of HIV at birth. Definitions: "in utero transmission" = +ve PCR result < 7 days of age. "peripartum transmission"
= +ve PCR result 7 days of age.
c Postnatal ARV Regimen: AZT is recommended, regardless of the mothers therapy. Zidovudine (AZT syrup,10 mg/mL), 4mg/kg/dose B.D.
to start within 8 hours of delivery, given for 6 weeks. The benefit of adding 3TC to AZT postpartum for 6 weeks if mothers have been on
combination of antiretrovirals during pregnancy is unclear. If added, dose of 3TC solution (strength, 10 mg/mL) is 2 mg/kg/dose B.D. to
start within 8 hours of delivery, for 6 weeks. If nevirapine is indicated, 2 mg/kg, single dose, to be given within 3 days of birth. No
confirmed short/medium term adverse events associated with in-utero/postnatal exposure to AZT. Concern with mitochodrial toxicity after
AZT +/- 3TC exposure in-utero remains to be confirmed.
d Any positive virological test (PCR or virus culture) must be confirmed on a separate sample due to the potential for false +ve results.
PCR has equivalent sensitivity of detection as virus isolation but is available sooner (takes ~1 week) than viral culture (46 weeks).
Close to 100% sensitivity of detection at 3 months in non-breast fed infants.
Traditionally, seroreversion i.e. disappearance of passive (maternal) antibodies is documented before declaring a child uninfected.
Antibody testing after 6 months may be useful to document declining maternal antibodies titres in uninfected babies.16

22

H U M A N

I M M U N O D E F I C I E N C Y
T A B L E

V I R U S

Perinatal Transmission (PNT) Risk Estimates


by Selected Intervention Strategies
ANTIRETROVIRALS
Mother
Pregnancy

Mother
Intrapartum

Baby

Delivery

Feeding

Vaginal

Formula

20%

Vaginal

Breast

40%

Vaginal

Breast

~36%

Caesarean

Formula

10%

(Full course AZT, 076 Study)

Vaginal

Formula

8%

19998

Caesarean

Formula

2%

Baseline(a)
Dunn, 19929
Nduati,

200010

(Uganda)(b)

The International Perinatal HIV Group,


199413

Connor,

The International Perinatal HIV Group,


Shaffer,

199917

19998

Vaginal

Formula

10%

Wade,

199818

Vaginal

Formula

10%

Wade,

199818

Vaginal

Formula

20%

NVP

NVP

Vaginal

Formula

~10%

Guay,

199912

(Short course AZT, Thai study)

PNT Risk

(HIV NET 012, Uganda)

Key: Shaded rows represent risks when no intervention strategies are in place. 0 = no ARV prophylaxis, + = ARV given. Unless stated, the
antiretroviral referred to is AZT. NVP = nevirapine
a
Prior to the introduction of recommended intervention strategies, a child born to an "asymptomatic" HIV infected woman with CD4+
counts > 200c/mm3 who did not breast feed had ~20% of being infected perinatally (in developed countries). For the purpose of these
algorithms, this figure is referred to as the "baseline" and increases or decreases in risk are referable to this baseline.
b
The approximate doubling of risk in breast fed babies over formula fed babies in this randomised clinical trial was seen over a range of
breast feeding duration (6 weeks to 2 years), with 75% infected in the first 6 months of breastfeeding.

T A B L E

GENERAL PRINCIPLES OF ARV IN PREGNANCY


1. ARV regimens are either treatment schedules or regimens
for preventing perinatal transmission (prophylaxis).
Women are now generally on HAART at time of pregnancy.
2. Pregnancy should not preclude use of optimal therapeutic
regimen.3 If initiating therapy, consideration could be given
to waiting till the second trimester. A possible association of
pre-term delivery with combination therapy has been found
but not confirmed. http://hivatis.org/guidelines
3. Efavirenz and hydroxurea (teratogenesis, animal studies)
are not recommended in pregnancy. Caution has been
raised with d4T + ddI in combinations with protease
inhibitors in pregnancy (fatal lactic acidosis. Pharmaceutical
company drug alert, January 2001)
4. The three part ZDV prophylactic regimen (Protocol 076) is
considered best practice, and is recommended for all pregnant women with HIV infection regardless of RNA viral
load.13 (Appendix 1) Thus, ZDV should be included as a
component of antenatal regimen where possible. If ZDV is
not included antenatally, intrapartum and newborn ZDV are
still recommended. However, if d4T is part of antenatal
ARV regimen, intrapartum ZDV is not recommended
because of potential drug interaction.

Suggested Testing Regimen


TESTS
Time

T Cell
Subsets

(Day 1)

PCR

Virus
HIV
Isolation Antibody

(+)

Week 1

Week 6

3 months

(+)

6 months

(+)

(+)

12 months

18 months
(if still seropositve at 12 mo)

Texts in parenthesis denote optional time or type of test.

APPENDIX 1: PACTG 076 ZIDOVUDINE REGIMEN13


Regimen
Time of ZDV
Administration
Antepartum
Intrapartum
Postpartum

Oral administration of 100 mg ZDV 5 times daily*, initiated at 14-34 weeks gestation and continued throughout the pregnancy.
During labour, intravenous administration of ZDV in a one-hour initial dose of 2 mg/kg body weight, followed by a continuous infusion of 1 mg/kg body weight/hour until delivery.
Oral administration of ZDV to the newborn (ZDV syrup at 2 mg/kg body weight per dose every six hours) for the first six
weeks of life, beginning at 8 -12 hours after birth.** (Note: intravenous dosage for infants who cannot tolerate oral intake
is 1.5 mg/kg body weight intravenously every six hours).
* Oral ZDV administered as 200 mg three times daily is an acceptable alternative regimen
** 4 mg/kg per dose, twice daily is an acceptable alternative regimen

23

Emendation 2006:
HUMAN IMMUNODEFICIENCY VIRUS
Nevirapine toxicity in women: The risk of hepatic toxicity is increased in
women particularly if CD4 counts are > 250 c/ml, usually in the first 6 weeks
of starting therapy. Pregnancy may be an added risk factor. Thus, caution is
warranted and close monitoring recommended if this agent is included in
therapeutic/prophylactic HAART regimens started during the antenatal period.
E10

Erratum, Table 1: In the Guay 1999 study (HIV NET 012), infants were breast
fed (not formula fed)

Revised suggested postnatal testing times and review


*HIV PCR at 1, 6, 12 weeks and at 6 months. Virus isolation and Tcell subsets are no longer routinely done in this setting.
If all PCRs remain negative, then clinical review only is recommended
at 12 months. HIV antibody at 18 months to document sero-reversion
builds further confidence in the diagnosis of non-infection in the
infant.
Nb: * As the proviral DNA PCR is currently not commercially available (2006), HIV
RNA PCR is used.

L I S T E R I A
A L G O R I T H M S

&

Diagnosis of Suspected Listeriosis and


Management of Proven Maternal Infection
Unwell febrile pregnant women
(includes flu-like illness)

Blood cultures
Gram stain and cultures
of genital tract
amniocentesis
Gram stain and culture

Positive for
Listeria monocytogenes

Negative

Consider other infections


and empiric antibiotics

Serious infection
including amnionitis

Mild
infections

Amoxycillin/Ampicillin
(46 g/day IV) and
Gentamicin for 14 days1, 2

Amoxycillin/Ampicillin
(23 g/day po)
for 14 days1, 2

Urgent delivery depending


on severity of maternal illness
and gestation

NOTES
Maternal listeriosis in 2nd/3rd trimester results in a mortality of 4050% for the fetus
Serology is not a useful tool for diagnosing listeria3
Early treatment of maternal infection can improve perinatal outcome
The recommended treatment regimens above are based on observations and case
reports. No randomized controlled trials have been performed to establish optimal
treatment regimens or to support efficacy of penicillin over ampicillin, but ampicillin or
amoxycillin is generally considered the preferred agent.13
Synergism exists for penicillin or ampicillin with gentamicin.3
Trimethoprim/sulphamethoxazole is suggested as an alternative in the penicillin allergic
patient.

24

L I S T E R I A
A L G O R I T H M

Antenatal Prevention Strategies


for Perinatal Transmission of Listeria
Pregnant women

Avoid
High Risk Foods

Use Safe
Food Handling Practices

Unpasteurized milk or food made


from raw milk

Thoroughly cook raw food from


animal sources

Pate, dips and soft cheeses

Chilled precooked seafoods

Keep uncooked meat separate


from vegetables, cooked foods
and ready-to-eat foods

Precooked meats and meat


products which are eaten without
further cooking or heating

Eat freshly cooked foods.


Avoid eating dips and salads in
which raw vegetables may have
previously been dipped

Thoroughly wash raw fruit


and vegetables

Reheat left-over or ready-to-eat


food until steaming hot

Uncooked or smoked seafood

Pre-prepared salads and coleslaws

Past history of listeriosis:


No role for vaginal cultures

NOTES
* Intrapartum antibiotic therapy is not recommended for mothers with a past history of perinatal
listeriosis. No data to suggest reculture during subsequent pregnancy has any value
* Asymptomatic vaginal carriage of L. monocytogenes is rare. Faecal carriage of L.
monocytogenes is found in 0.616% of the population at any one time.4 The significance of
faecal excretion in perinatal infection is uncertain.
* If listeria found on vaginal/rectal culture, it may be appropriate to try to eradicate the
organism before delivery with oral amoxycillin or erythromycin (250 mg q6hrly for 10 days).
While this approach may seem reasonable it has not been definitively studied.5
* Local Public Health Department publications, with detailed advice, are available.

25

L I S T E R I A
A L G O R I T H M

Diagnosis and Management of


Infant at Risk of Perinatal Listeriosis
Maternal listeriosis
(proven or suspected)

Still
birth

Unwell neonate
Suspicious clinical findings:
placental, cord or postpharyngeal granulomas
Multiple small skin
granuloma, papular or
pustular skin rash
Meconium stained liquor
<34 weeks gestation
Purulent conjunctivitis

Well
neonate

NOTES
Mortality rates from 350% in
infected neonates born alive7

Perinatal listeria can present as


early-onset disease (within 7 days
of birth) often associated with
prematurity and fulminant disease,
and late onset disease (7 days to
6 weeks), often presenting with
meningitis.
Surface cultures with Gram stain
from placenta, meconium, rectal
and external ear canal have all
been found to have a high yield in
isolating the organism.5, 8
Optimal antimicrobial therapy for
various manifestations of listeriosis
has not been established in
controlled clinical trials and
remains controversial. No
controlled trials available to
establish a drug of choice or
duration of therapy.13
Alternative antibiotics:
Trimethoprim/sulphamethoxazole
reserved in the of event of lack of
response to standard therapy;
Rifampicin effective in vitro but
inadequate clinical information
available; erythromycin sensitive
but bacteriostatic; no role for
cephalosporins.

Septic workup
Blood cultures, CSF
Superficial cultures
with Gram stain
Culture placenta
CXR, Urine culture
FBC

Empiric treatment:
Amoxycillin/ Ampicillin (50 mg/kg q12hrly)
and gentamicin (2.5 mg/kg q12hrly)6,7

Culture positive or unwell at


diagnosis: continue antibiotics
CSF positive:
Amoxycillin/Ampicillin and
gentamicin 21 days
CSF negative:
Amoxycillin/Ampicillin and
gentamicin 14 days

26

Well neonate and


culture negative:
Stop antibiotics at
48 hours

M Y C O B A C T E R I U M

T U B E R C U L O S I S

A L G O R I T H M

Antenatal Diagnosis
Symptoms
suggestive
of TB

HIV
positive

Close contact of
infectious TB

Recent arrival from area


with high prevalence of TB

Mantoux test

Negative

Positive

No further action

High clinical suspicion

Examine for signs of TB +


Perform chest X-ray*

No evidence
of TB

Evidence of old
pulmonary TB

Evidence of
active TB

Mantoux conversion
> 2 years or unknown

High risk** or Mantoux


conversion within previous
2 years

Sputum, urine +/- other


specimens, or investigations
as appropriate

Reassess need for INH


prophylaxis post partum

INH prophylaxis from


2nd trimester
ALGORITHM 2

* Chest X-ray may be omitted if the risk of active TB is considered to be low.


** High risk HIV positive, those with medical conditions that increase the risk for reactivation of inactive TB, eg diabetes, chronic renal
failure, malignancy.
NOTES
The development, clinical presentation and progression of TB
are not altered by pregnancy.1,2
Although data are conflicting, pregnancy is not thought to
increase the risk of inactive TB becoming active.
The symptoms of extrapulmonary TB are frequently non-specific,
and may be attributed to physiological changes of pregnancy.
Areas with high prevalence of TB include South East Asia,
Pacific Islands, Africa, Eastern Europe, Latin America.
Mantoux testing of contacts is usually performed by local Health
authorities, and may need to be repeated at 12 weeks after
break of contact.
Mantoux skin testing: intradermal injection of 0.1 mL of a 100
international unit/mL solution of purified protein derivative (PPD),
with induration measured at 48-72 hours.
The Mantoux test is not affected by pregnancy.
Chest X-ray should be performed with appropriate abdominal
shielding.

Isoniazid (INH) "prophylaxis" is not true prophylaxis, and is often


referred to as "treatment of latent TB infection". Usual duration is
6 months.
INH is safe in pregnancy.3
Pyridoxine should be given with INH to pregnant and breastfeeding women (50 mg/day), and to their breast-fed infants (10
mg/day) whether or not the infant is taking INH.4,5
Interpretation of Mantoux skin test positivity [Source: American
Thoracic Society, 1999]
5 mm diameter in people with HIV infection, in people in close
contact with someone with infectious TB, or in people with a
chest X-ray suggestive of previous TB;
10 mm diameter in recent arrivals (< 5 years) from high prevalence areas, in injecting drug users, residents/employees of prisons, homeless shelters, residential facilities for AIDS patients,
high risk patients** or in children < 4 years old;
15 mm diameter in those 4 years old with no risk factors for
TB, and in children who have had BCG.

27

M Y C O B A C T E R I U M

T U B E R C U L O S I S

A L G O R I T H M

Initial Management of
Suspected Maternal TB
Proven maternal TB

Low risk of
INH resistance

INH
Rifampicin
Ethambutol
Use of Pyrazinamide is optional*

High risk of
INH resistance

INH
Rifampicin
Ethambutol
Pyrazinamide

9 months
9 months
2 months

6
6
2
2

months
months
months
months

NOTES
Active TB during pregnancy must be treated immediately. This is true for cases in which TB has not been confirmed,
but is considered likely on clinical grounds.
TB does not affect the course of pregnancy or type of delivery required.1,2
High risk of INH resistance should generally be assumed, particularly for HIV-positive women, recent arrivals from an area of high
prevalence, and those who have had previous anti-TB treatment.
Duration of therapy with each drug may vary according to the resistance pattern of the isolate, and according to the form of TB
(e.g. longer for TB meningitis).
*The duration of treatment with INH and Rifampicin is longer for cases when Pyrazinamide is not given in the first 2 months.
Directly observed therapy (DOT) is ideal practice, but may not be feasible for all patients with TB (the states vary on the application of
DOT).
All of the anti-TB drugs cross the placenta and reach a low concentration in fetal tissues.3 However, INH, Rifampicin and Ethambutol
are all safe in pregnancy. Little is known about the effects of Pyrazinamide in pregnancy, but it has been used without adverse
effects. Many internationally recognised TB organisations recommend its routine use in pregnancy.6 Streptomycin is contraindicated
in pregnancy.6
The risk of INH-induced hepatotoxicity appears to be higher in women, and may be more so in the perinatal period. Women should
be monitored for hepatotoxicity with monthly ALT/AST.7
300 mg po daily (give with Pyridoxine 50 mg daily note increased dose in pregnant and

INH
breastfeeding women).

450 mg po daily (< 50 kg)


Rifampicin
600 mg po daily ( 50 kg).

15 mg/kg po daily.
Ethambutol
Pyrazinamide

1.5 g po daily (< 50 kg)


2 g po daily ( 50 kg).

28

M Y C O B A C T E R I U M

T U B E R C U L O S I S

A L G O R I T H M

Management of the Neonate


Maternal TB is likely
to be associated with
haematogenous spread*

Active pulmonary TB
mother infectious
at time of delivery

Assess neonate
for clinical evidence
of congenital TB

Mother on
anti-TB treatment
not infectious

Mother completed
anti-TB treatment
not infectious

Assess neonate
for clinical evidence
of congenital TB

Assess neonate
for clinical evidence
of congenital TB

Absent

Present

Absent

Chest X-ray and


gastric aspirates x 3

Chest X-ray, gastric aspirates


x 3 and lumbar puncture

INH for
6 months

Mantoux test
at 3 months
and 6 months

No evidence
of TB

TB

INH, Rifampicin, Pyrazinamide


+ Amikacin or Ethionamide

INH for
6 months

Positive at
any time

Investigate according to clinical


state and treat as above

Positive at
any time

Mantoux test
at 3 months
and 6 months

Negative at
6 months

BCG should be given if there is


any possibility of future
exposure to TB

Negative at
6 months

Absent

Mantoux test
at 3 months

* Disseminated or miliary TB, tuberculous meningitis, etc. The placenta or maternal genital tract may become infected, and congenital
infection may ensue. However, congenital TB remains very rare.7,8
Sputum smear positive

NOTES
Most cases of neonatal TB occur as a result
of airborne spread after delivery. However,
separation of mother and neonate is only
necessary if the mother is sick enough to
require hospitalisation for TB.5
Other family members and close contacts
should be assessed for TB infection or
disease. If a close contact is infectious,
separation is preferable, but, if impossible,
INH prophylaxis should be given until the
contact has been culture-negative for 3
months.
Respiratory distress,hepatosplenomegaly,
fever, lymphadenopathy and poor feeding
are the most common presenting features
of congenital TB.8,9
If congenital infection is suspected, the
placenta should be examined and
microscopy, culture and histology performed.

susceptibility results are available.


The Mantoux test is likely to be negative for
the first few weeks of life, even if the
Ethionamide or Prothionamide 1520
neonate has TB.3,4,10
mg/kg daily until drug susceptibility results
are available. May be difficult to obtain.
Mantoux conversion may be delayed for up
to 6 months; thus INH prophylaxis must be Ethambutol 15 mg/kg po daily may be used
continued until this time.
in place of Amikacin or Ethionamide, but
should be reserved for special cases. It
Drug treatment
may induce optic neuritis, which is difficult
The decision regarding number and choice
to identify in infants.
of drugs for management of neonates and
Streptomycin 1520 mg/kg im daily may
infants with TB is difficult, and warrants
also be used in place of Amikacin or
specialist advice.
Ethionamide, but injection is painful and it
INH 5-15 mg/kg po daily for 6 months
is difficult to obtain.
(Pyridoxine 10 mg po daily must be added
These drugs are excreted in breast milk.
for breastfed infants).
If a breastfeeding mother and neonate are
Rifampicin 1020 mg/kg po daily for 6
both on anti-TB therapy, there is a small
months.
risk of toxic levels in the neonate. This can
Pyrazinamide 1530 mg/kg po daily until
be minimised if the mother takes her
drug susceptibility results are available.
medications immediately after a breast
Amikacin 15 mg/kg iv daily until drug
feed.

29

P A R V O V I R U S
A L G O R I T H M

Parvovirus B19 Infections During


Pregnancy: Risk Assessment
EXPOSURE DURING EPIDEMIC

Risk of infection if susceptible


after exposure at home
is up to 50%1

60% of women of childbearing age are immune


to parvovirus infection1,3

Risk of infection if susceptible


after exposure at school or
child care = 2030%1,2

Risk of infection if susceptible


after exposure in community
is up to 20%

40% of women of child-bearing age are


susceptible to parvovirus infection3

RISK OF INFECTION IN PREGNANCY

50% x 40%
20%

2030% x 40%
= 812%

20% x 40%
8%

3% hydrops (between 9
and 20 weeks gestation)4,5

<1% (no excess)


congenital abnormalities4,5

OUTCOME
(PROVEN MATERNAL
INFECTION)5,6,7
10% excess fetal loss in
first 20 weeks of pregnancy4

32% spontaneous
resolution (usually
within 8 weeks)

NOTES
It is not practicable to prevent
exposure at home. Exclusion from
work of pregnant school teachers
or child care workers is not
recommended during a parvovirus
epidemic (nor is exclusion of
infected children).
Susceptible pregnant HCW should
not care for patients with known
complicated parvovirus infection (eg
aplastic crisis) or chronic parvovirus
infection (eg in an
immunocompromised host).
Routine antenatal screening is not

33% death without IUT


(usually within 45 days of
first abnormal ultrasound)

indicated.
There is a 50% risk of transmission
from an infected mother to her
fetus.8
Fetal loss = 15%, compared with
5% overall (ie excess loss = 10%).4
Onset of hydrops 2-17 weeks
(average 5 weeks) after maternal
infection.
Congenital abnormalities
anecdotal reports only (less than
rate of major malformations in
newborns of 2%).4,5
IUT = intrauterine transfusion.

30

27%
resolution
after IUT

6%
death after
IUT

OVERALL RISKS
Pregnant woman with
Any pregnant woman
exposed to parovovirus proven recent infection
Excess fetal
loss in first
20 weeks
Death from
hydrops
or its
treatment

0.41%
(1 in 1001 in 250)

5%
(1 in 20)

0.050.1%
(1 in 8501 in 2000)

0.6%
(1 in 170)

Pregnant women who are exposed should be informed


of risks, and offered serological testing.

P A R V O V I R U S
A L G O R I T H M

Parvovirus B19 Infections During Pregnancy:


Antenatal Diagnosis and Management
DIAGNOSIS OF INFECTION FOLLOWING CONTACT OR ILLNESS
WITH RASH DURING PREGNANCY9,10
(Check rubella serology symptoms are similar and cross-reactions can occur)

SEROLOGY

INTERPRETATION

ACTION

IgG +
IgM

IgG
IgM

IgG
IgM +

IgG +
IgM +

Immune

Susceptible

? Recent
infection*

Recent
infection

Nil

Repeat IgG 24 weeks after exposure


or if symptoms occur

IgG
(false positive IgM)

IgG +
(recent infection)

See
ALGORITHM 3

NOTES
IgM is detectable within 13 weeks of exposure and usually remains
detectable for 23 months.
IgM reference standard: mu-capture radioimmunoassay (MACRIA).
Commercial IgM test kits (EIA or IF):
sensitivity: 70-80% overall (100% in adults with arthropathy; lower in children)
specificity: 92-97% overall (70-85% in patients with other infections, including rubella)9
Note: absence of IgM does not exclude recent infection
* Symptoms include non-specific illness, rash, and/or arthralgia/arthritis).

31

P A R V O V I R U S
A L G O R I T H M

Parvovirus B19 Infections During Pregnancy:


Management of Proven Maternal Infection
RECENT MATERNAL INFECTION

Ultrasound examinations at
12 week intervals for 612 weeks
after maternal infection5

Normal

Fetal hydrops

No
further action

Refer to specialist experienced


in fetal ultrasound, blood
sampling and transfusion

Fetal blood
sampling

Daily
ultrasound

Resolution

Mild anaemia, normal


platelets, reticulocytosis

Moderate to severe anaemia


+/- thrombocytopenia

IUT

Fetal blood
sampling

Progression

NOTES
No intervention is available to prevent fetal infection or damage.
Termination is not indicated because of low risk of fetal damage.
Amniocentesis for diagnosis of asymptomatic intrauterine fetal
infection is not recommended.
a-fetoprotein levels are not helpful previous reports that
increased levels predict poor outcome have not been confirmed.5
Pregnancy should be monitored by repeated ultrasound

examination to detect hydrops fetalis.


A fetus with mild hydrops may be profoundly anaemic.
Fetal blood sampling measure haemoglobin, platelets and
reticulocyte count.
No specific investigation is indicated in normal infants.
Infants in whom hydrops has occurred and resolved should be
monitored for evidence of anaemia.

32

R U B E L L A
A L G O R I T H M

Diagnosis of Suspected
Maternal Rubella Infection
Indications for testing pregnant women

a) Routine antenatal screening (IgG only)*,1,2


If IgG ve, immunize after delivery
If IgG +ve; if >10 IU/mL; minimal risk of reinfection
If <15 IU/mL; consider reimmunisation after delivery

b) Rubella testing (IgG/IgM)** because of


(i) contact with rubella
(ii) rubella-like illness (fever, erythema, arthralgia)

IgG +ve
IgM +ve
Possible recent infection
(or reinfection,
depending on history)

Repeat to
confirm

IgG ve
IgM ve
Susceptible

IgG ve
IgM +ve
Possible recent infection

Repeat if <3 weeks


since contact or <7days
since onset of illness

Repeat

IgG seroconversion***

No seroconversion

Retest by EIA or HAI


on IgM fractions
after sucrose density
gradient centrifugation

Susceptible
(possible false
positive IgM)

Positive
IgM
confirmed

Counsel and manage as per ALGORITHM 2

Immunise after delivery

33

IgG +ve
IgM ve
Past infection
or immunisation;
manage as for positive
antenatal screening

NOTES
*Different laboratories
use various cut-offs for
reporting low IgG levels
ranging from 7 to as high
as 50 IU/mL. Levels
corresponding to
protection from
reinfection are
imprecise, but only a
small proportion of
women are affected by
reinfection.1,2
** Reinfection can occur
without detectable IgM.
Previously stored serum,
if available, should be
retrieved and tested in
parallel with current
serum, for evidence of
pre-existing antibodies
or seroconversion.
*** Seroconversion
should be checked by
testing the sera in
parallel.

R U B E L L A
A L G O R I T H M

Management of Proven
Maternal Rubella Infection
Maternal infection*

Primary infection

Reinfection

Risk of fetal damage or


congenital rubella syndrome (CRS)
related to timing of maternal infection59**

< 8 weeks
90100%

812 weeks
50%

If asymptomatic reinfection with a reliable


history of previous positive serology, then
risk of fetal damage is <5%3,4
If typical clinical rubella or doubtful
previous immunity, risk must be assumed
to be the same as for the first infection

1220 weeks
20%

>20 weeks
< 1%

Consider termination of pregnancy if maternal infection in first trimester.


If maternal infection occurred in second trimester, consider fetal testing.

Prenatal fetal diagnosis/testing


Rubella PCR, rubella culture and fetal IgM can be performed following
chorionic villus sampling (CVS)/amniocentesis or cordocentesis9,10 ***
BUT
CVS is associated with risk of contamination with maternal tissue
giving false +ve PCR
PCR is not widely available and sensitivity is generally not well
validated. However, a positive result will be helpful
(assuming that contamination can be excluded)11
False negative fetal IgM is common until late in pregnancy 10,12

NOTES
* No specific management of mother (rubella specific immunoglobulin not available and normal human immunoglobulin NOT indicated).
** Transmission risks and details of incidence and type abnormalities can be found in textbooks8,9
*** Contact your local Public Health Virology Laboratory for information about the availability of rubella culture or PCR

34

R U B E L L A
A L G O R I T H M

Management and Follow Up


of the Infant at Risk of Infection
At birth
Ensure clinical attendants are rubella vaccinated and have specific
antibodies detected
Examine infant for evidence of CRS (growth retardation, eye/cardiac
abnormalities, rash, haematological abnormalities, pneumonitis,
osteitis are most likely to be detectable at birth)
Investigate infant
Serology (IgM)
PCR (urine and throat swab); note -ve PCR does not exclude infection
Culture urine and throat swabs, tears (conjunctival swab),
lens tissue (if available); results can take several weeks

Clinical features CRS


IgG maternal IgG titre
(measured in parallel)
IgM +ve
PCR +ve

No clinical features CRS


IgM +ve
PCR +ve

No clinical features CRS


IgG maternal IgG titre
(measured in parallel)
IgM ve
PCR ve

Symptomatic, infected infant

Asymptomatic, infected infant


(risk of late onset disease
months or years after birth)

Infant probably not infected

No specific management

Breastfeeding not contraindicated

Ensure ophthalmology, cardiac and hearing assessments at birth

Regular assessments (3 to 6 monthly) necessary in the first few


months and years of life to detect the emergence of
abnormalities related to persisting infection (deafness,
neurological deficiencies, epilepsy, cataracts, retinopathy, tooth
defects and growth retardation)13
Infants infectious for at least 12 months after birth and are a
hazard to susceptible female staff and pregnant contacts

Infant should be isolated (droplet and contact) while in hospital

Ensure all hospital contacts/caregivers are rubella immune

35

Reassure
Breastfeeding not
contraindicated
Confirm absence of infection
with falling/absent IgG at or
after 9 months of age

S T R E P T O C O C C U S ,
A L G O R I T H M

G R O U P

Management of Pregnancy with Respect


to Group B Streptococcal (GBS) Infection
Strategy A*
Prenatal screen for maternal GBS
colonisation

Strategy B
Obstetric
Risk factors alone

Screen at 3537 weeks gestation


Low vaginal + anorectal swabs **
Selective broth medium

GBS
colonisation

GBS
negative

GBS unknown (failed


screening, refused swab or
result unavailable)

Obstetric risk
factor(s)***

No chemoprophylaxis
unless previous infant
with GBS infection or
GBS bacteruria
(current pregnancy) of
any colony count.

Yes

Intrapartum chemoprophylaxis
(SEE ALGORITHM 2)

Obstetric risk factors ***


Previous infant with EOGBS
GBS bacteriuria (any colony count) this
pregnancy
Spontaneous onset of labour
< 37 weeks gestation
Rupture of membranes 18 hrs
Intrapartum fever 38oC

Nil

One or more
present

No

No chemoprophylaxis

Intrapartum chemoprophylaxis
(SEE ALGORITHM 2)

** Detection of GBS is increased by up to 25% by collecting an


anorectal swab in addition to a vaginal swab.6 A single swab may
be used, provided the vagina is swabbed prior to the anorectal
area. Samples may be obtained by the patient.
Maternal carriage of GBS does not predict premature rupture of
membranes or preterm delivery.7
Selective broth media are more sensitive than standard solid
media. Examples include NPC broth (Todd-Hewitt broth
supplemented with colistin, nalidixic acid and crystal violet) and
semi-solid new Granada medium.8
*** The obstetric factors listed are associated with increased risk
for EOGBS.2,9 However, 25-30% of cases are not associated
with maternal risk factors.2
Babies born to women with GBS bacteriuria (any colony count)
during pregnancy are more frequently and more heavily
colonised with GBS, increasing the risk of EOGBS.

NOTES
Colonisation of the genital tract with GBS occurs in
approximately 20% of women. Early onset GBS disease
(EOGBS, within the first week of life) occurs at a rate of 12 per
1000 live births, although this is declining.1
Intrapartum chemoprophylaxis is highly effective in reducing
neonatal colonisation with GBS and preventing EOGBS.1,2,3
* A recent Centers for Disease Control & Prevention (CDC) study
in >600,000 live births found the Screening approach >50%
more effective than the Risk Factor approach in preventing
GBS disease.4 The application of this in Australia remains to be
determined.
Prenatal screening (<35w) to detect GBS does not reliably
predict carriage at delivery. The later in pregnancy that cultures
are performed, the better the correlation with culture results at
delivery (particularly within 5 weeks of delivery).1,5,6

36

S T R E P T O C O C C U S ,
A L G O R I T H M

G R O U P

Intrapartum Antibiotic Prophylaxis for


Prevention of Early Onset Neonatal GBS Sepsis
GBS +ve, GBS -ve but previous infant with GBS
sepsis, GBS bacteruria (this pregnancy) or GBS
unknown with obstetric risk factors

GBS -ve or GBS unknown with no obstectric risk


factors

Signs of
maternal
sepsis

Signs of
maternal
sepsis

No

Give intrapartum chemoprophylaxis


Benzyl penicillin 1.2 g IV loading dose,
followed by 0.6 g IV 46 hourly, from
onset of labour until delivery

If penicillin hypersensitivity:
clindamycin 600 mg IV 8 hourly
OR
erythromycin 500 mg IV 6 hourly

Yes

Yes

No

Collect

Endocervical swab

Urine (m/c/s)

Blood cultures

Full blood count

CRP

Give broader spectrum antibiotics


amoxycillin 2 gm IV 6 hourly
+
gentamicin 46 mg/kg IV daily
+
metronidazole 500 mg IV 12 hourly

No antibiotics

NOTES
90% of neonates with EOGBS have onset of signs within 12 hours of birth (suggesting intrauterine transmission), so intrapartum antibiotic
prophylaxis is the most effective means of prevention.
The rate of fatal maternal anaphylaxis to penicillin is estimated at 1 in 100 000. Less severe reactions occur in 710%.
Clindamycin and erythromycin are active in vitro against GBS, but their efficacy in prevention of EOGBS has not been evaluated.
Clindamycin resistance has been reported in 3.4% of invasive GBS isolates, and erythromycin resistance in 7.4% (USA). Nonetheless,
they are recommended by most experts as alternatives for women who have hypersensitivity to penicillin.6
Pathogens responsible for chorioamnionitis include GBS, anaerobic cocci, and enteric Gram-negative bacilli (often polymicrobial).

37

S T R E P T O C O C C U S ,
A L G O R I T H M

G R O U P

Management of Infant Born to Mother


Who Received Intrapartum Prophylaxis
Signs of sepsis

Yes

No

< 35 weeks gestation

35 weeks gestation

FBC, BC, urine and


CSF cultures, CXR

FBC, BC

Adequate IP

CSF suggestive of
meningitis

Observe in hospital
for 48 hours

Yes

Benzylpenicillin
+
Cefotaxime

No

No

Signs of sepsis
at any time

Benzylpenicillin
+
Gentamicin

Yes

Observe in hospital
for 48 hours

ANTIBIOTIC DOSES
Benzylpenicillin 50 mg/kg IV
12 hourly 1st week of life
over 1 week of age
6 hourly

NOTES
The relative risk of EOGBS is significantly greater in preterm neonates than
in babies born at term
Duration of intrapartum prophylaxis (IP) is inversely proportional to the
percentage of babies colonised with GBS
Adequate IP is defined as 2 doses of antibiotics given before delivery
FBC = full blood count; BC = blood culture;
CSF = cerebrospinal fluid; CXR = chest X-ray
Urine must be obtained by suprapubic aspiration or urethral catheter
IP = intrapartum (antibiotic) prophylaxis
Caftriaxone should be avoided in neonates.

38

Cefotaxime 50 mg/kg IV
12 hourly
preterm
8 hourly
1st week of life (term)
6 hourly
over 1 week of age
Gentamicin single daily dose
2.5 mg/kg < 30 weeks gestation
3.5 mg/kg 3035 weeks gestation
5 mg/kg
1st week of life (term)
7.5 mg/kg 1 week10 years of age

T O X O P L A S M A
A L G O R I T H M

Antenatal Evaluation
Routine antenatal screening not
generally recommended in Australia
but is sometimes donea

Serology done because of symptoms


? acute toxoplasmosis: malaise, fever,
lymphadenopathy (eg cervical)

Serology result

IgG ve
IgM -ve

No past infection
Education re: preventionb
Repeat if symptomatic
or routine screeningc

IgG +ve
IgM ve

IgG +ve
IgM +ve

Past infection
No further action

Asymptomatic and
inconsistent/low
+ve IgM
IgA ve and/or
high IgG avidity

Possible recent
infection

Check symptoms and


repeat IgM (different kit)
+/or IgA
+/or IgG titre
+/or IgG avidity d

Negative

Seroconversion
Positive

NOTES
a. Pros & cons of antenatal screening
complex; if it is done, there should be
an appropriate management plan.
b. Avoid raw/undercooked meat; wash
hands after gardening; wash raw
vegetables etc.
c. Various protocols recommend repeat
testing after 1-6 months or at delivery,
to identify seroconversion.
d. IgM can remain +ve for months or
years; IgA, rising IgG level and/or low
IgG avidity are more specific for
"recent" infection (within ~3 months)

Recent toxoplasmosis
definite or probable

ALGORITHM 2

39

Symptoms and/or
additional testing
consistent with recent
toxoplasmosis
Repeated/high +ve IgM
+ve IgA
low IgG avidity

T O X O P L A S M A
A L G O R I T H M

Investigation and Management


of Maternal Toxoplasmosis
Evidence of recent acute maternal
toxoplasmosis (see ALGORITHM 1)
Seroconversion and/or
IgM +ve; IgA +ve; low IgG avidity
symptoms

Risk assessmenta depends on trimester1

Second Trimester:
Fetal infection intermediate risk
(25-40%); if infected, damage
intermediate risk (15-25%)

First Trimester:
Fetal infection low risk (5-15%)a
if infected, damageb high risk
(60-80%)

Third Trimester:
Fetal infection high risk (30-75%)
if infected, damage low risk
(2-10%)

Antibiotic therapy
Consider treatment of mother at diagnosis (depending
on gestation and certainty of diagnosis).
+ve (1st trimester)
consider termination

Intrauterine diagnosis2,3
ultrasonography
amniocentesis d at 18-20 wks gestation or
4 weeks after maternal infection

-ve fetus not


infected; continue
therapyc if maternal
infection fairly certain
NOTES
a. Estimated risks also vary according to the
methods of diagnosis, duration of followup and treatment.
b. Classic signs of congenital toxoplasmosis
are: chorioreinitis; hydrocephalus;
intracranial calcification.
c. Depending on certainty of diagnosis.
Treat with spiramycin (difficult to obtain in
Australia) or
pyrimethamine/sulphadoxine (potentially
toxic in 1st trimester; use with folinic
acid); efficacy of treatment has not been
confirmed in randomised controlled
trials.5,6
d. For PCR and/or culture (sensitivity
~80%); no additional benefit from fetal
blood testing.
e. Neonatal screening and early treatment of
infant is an alternative to antenatal
screening.

+ve (2nd or 3rd trimester)


consider termination if ultrasound
abnormal or
treat mother with sulfadoxine & pyrimethamine (& folinic acid) continuously or
alternating with spiramycin until delivery.
confirm diagnosis in infant

Postnatal investigation and


management of infant at risk

40

Neonatal screeninge4

ALGORITHM 3

T O X O P L A S M A
A L G O R I T H M

Investigation and Management of


Infant at Risk of Toxoplasmosis
Neonatal screen +vea

Full clinical examination


including ophthalmological
and cerebral ultrasound

Maternal infection
treated or untreated

IgM or PCR positive;


+/or infants IgG titre significantly
more than mothers

Investigations:
Infant blood IgM +/or
IgA;4 persistent IgG;
placental
histology/PCR; blood
or CSF PCR

Negative

Clinical and placental


examination normal
& IgM -ve

Symptomatic congenital toxoplasmosis


(minority of infected infantsb)
chorioretinitis/retinal scarring;
intracranial calcification; hydrocephalus;
hepatosplenomegaly; pneumonia;
thrombocytopenia; lymphadenopathy;
myocarditis & IgM +ve +/or abnormal
placenta +/- CSF abnormality (PCR +ve)

Positive result +/or IgG


persists >6 months

Repeat IgG at
6 months of age

Negative

Positive

Infant not infected;


no further action

Asymptomatic congenital toxoplasmosis


(majority of infected infants)

Treatc infant for 12 months with


pyrimethamine & sulphadoxine
(plus folinic acid)
continuously or alternating with spiramycin
(varying combinations used)

NOTES
a. Neonatal screening not often done, but is an alternative to antenatal screening to detect infected infants for treatment.7
b. Proportion of infants infected and severity depends on when maternal infection occurred and if/how treated.
c. High incidence of longterm sequelae (eg chorioretinitis) in untreated infants even if asymptomatic at birth can be reduced by treatment.

41

T R E P O N E M A

PA L L I D U M
A L G O R I T H M

( S Y P H I L I S )

Routine Antenatal Screening


for Syphilis
Non-treponemal screening test
VDRL or RPRa

Positive

Specific treponemal
test e.g. TPPA &/or
FTA-Abs

Positive

High-risk subject

Negative

VDRL &/or RPR titre

Specific treponemal screening test


eg. TPPA, TPHA, syphilis EIA

OR

No

No further
testing

Negative

Positive

Yes

Negative

Repeat at 2832 weeks


gestation and at delivery

VDRL &/or
RPRa titre
& FTA-Abs

Negative
High risk subjectc or symptomatic
Biological
false
positive
VDRL
/RPRb

Both
positive

VDRL
negative FTAAbs positive

Both
negative

? False negative treponemal test


(rarely in very early infection)
Past treated/
latent
infection
Repeat in 4 weeks

Positive

Negative

Biological
false positiveb

False
positive
screening
test

SYPHILIS

Trace sexual contacts


and other children

Perform testing for other sexually


transmitted diseases (including
HIV)

NOTE:
Lettered superscripts refer to notes (see page 44). For reading list, see page 54.

42

Investigate and treat as per


ALGORITHM 2

T R E P O N E M A

PA L L I D U M
A L G O R I T H M

( S Y P H I L I S )

Investigation and Treatment of


Maternal Syphilis
Symptomatic syphilis (rare)
Tissue diagnosis:
(biopsy, exudate etc.)
Darkfield microscopy
Syphilis PCR
Fluorescent treponemal
antibody staining (DFA)h
Rabbit infectivity test

PRIMARY
Ulcer (chancre)
Anogenital
Oral/breast (rare)

Seropositive

Antenatal screening

DETERMINE STAGE
Clinical history/examination
Risk factors sexual contacts, other/previous STDs, IV drug use

SECONDARY
Systemic illness
Fever, rash, hepatitis, lymphadenopathy, meningoencephalitis

LATENT
Asymptomatic
Time since seroconversion (usually not
known): <2 years early; >2 years late

TERTIARY
Cardiovascular
Neurological
Granulomata

? CONGENITAL
Stigmata or
asymptomatic
No risk factors

RISK OF FETAL INFECTION


HIGH

MODERATE

Skin test

Reactive

? Treat
with
cefotaxime
(efficacy
unproven)

Yes

Nonreactive

Desensitise

LOW

NEGLIGIBLE

? Penicillin allergy

Treat with penicillin according to stage


Preferably IM procaine or IV benzylpenicillin
Benzathine penicillin if daily follow-up impossible; crosses
placenta poorly (see Therapeutic Guidelines: Antibiotic)

Repeat VDRL/RPR monthly (or


at delivery)

Negative, low or stationary within


6 months

Neurological signs and


symptoms

Titre persistently
raised >6 months

Lumbar
puncture

CSF
cell count
protein
VDRL &
specific
treponemal
test, PCR

Titre rising near term ?


reinfection

Successful treatment
12 weeks gestation

Successful treatment >12 weeks


gestation to 4 weeks prior to
delivery

Treatment <4 weeks prior to


delivery OR non-penicillin antibiotic
OR benzathine penicillin

Negligible risk of fetal damage

Fetal damage possible

Risk of congenital syphilis

Repeat
treatment

ALGORITHM 3

43

T R E P O N E M A

PA L L I D U M
A L G O R I T H M

( S Y P H I L I S )

Investigation and Management of a


Neonatej Born to a Mother with Syphilis
Mother adequately treated with penicillin (Algorithm 2)
>4 weeks prior to delivery 4 fold decrease in VDRL or
RPR titre (repeat maternal serology at delivery)

Yes

No

Congenital syphilis
possible

Infant unlikely to
be actively infected
Full clinical examination

Normal

Abnormale

Negativef

Positivef

INVESTIGATE
LFT, FBC, UEC; Bone scan
& long bone x-rays; CSFi cell
count, protein, VDRL, PCR, IgM

All investigations
negative
Not infected

Positive

Congenital syphilis symptoms


Abnormal CSF neurosyphilis

Serological follow-up: VDRL/RPR at 1, 2, 4, 6 and


12 months of age or until non-reactive on 2 occasions
Neurosyphilis: repeat CSF examination at 6 months

NOTES
a. VDRL has largely been replaced by RPR (except for CSF
examination). RPR titre is generally higher than VDRL and titres
cannot be directly compared.
b. Biological false positive VDRL/RPR results occur in people with
intercurrent viral and other infections, autoimmune diseases and in
pregnancy.
c. High risk groups: young age, single, low socioeconomic status,
poor education, poor antenatal care, substance abuse, prostitution.
d. Untreated maternal infection in the first trimester is more likely to
produce fetal damage.
e. Clinical abnormalities suggestive of congenital syphilis include rash
(maculopapular or vesicular), mucosal lesions, nasal discharge,
hepatomegaly, bony tenderness and eye lesions. If serology is
negative investigate for other causes.
f. Infant serology negative if VDRL/RPR titre at least four times less
than the mothers and EIA IgM negative; positive if VDRL/RPR
titre at least four times greater than the mothers and/or EIA IgM

Positive

Indeterminatef

Western blot

Benzyl penicillin 50 mg/Kg BD IV 10 days or


Procaine penicillin 50 mg/Kg daily IM 10 days

NO FURTHER
ACTION

Placental histopathology PCRh

Infant serology (VDRL &/or RPR,


EIA IgM)

Negative

g,h

Negative

Congenital syphilis unlikely


Repeat serology at 3 months
& 6 months; treat if serology
positive or follow-up
doubtful

Retreat if persistently
reactive serology, abnormal
CSF or clinical examination

positive; indeterminate if otherwise.


g. Western blot positive if infant serum reacts with additional bands
compared with mothers serum.
h. Western immunoblotting for syphilis antibodies, syphilis PCR and
placental darkfield microscopy, PCR and fluorescent
antitreponemal antibody staining are available as additional
confirmatory tests in reference laboratories.
i. If CSF examination is not possible then give full 10 day penicillin
course and, if possible, perform lumbar puncture at six months to
exclude persistent neurosyphilis.
j. Infants whose mothers develop secondary or early latent syphilis
within 1 year of delivery should be tested and treated if they have
positive serology.
k. The clinical presentation of late congenital syphilis (keratitis,
deafness, Hutchinsons teeth etc.) may be delayed for several
years and regular assessment may be warranted.
l. Serology should be performed on neonatal serum rather than cord
blood because of the risk of contamination with maternal blood.

44

V A R I C E L L A

Z O S T E R

A L G O R I T H M

V I R U S

Exposure to Varicella Zoster Virus


During Pregnancy
Previous maternal
chickenpox

No history or uncertain
past history of chickenpox

Check serology urgently

Serology not available

Seropositive

Seronegative

No action required

Assess time of exposure

Exposure
< 96 hours
earlier

Exposure
> 96 hours
earlier

Passive immunisation
ZIG

NOTES
Exposure to Varicella or Zoster
for which ZIG is indicated for susceptible persons7
Living in the same household
as a person with active
chickenpox or herpes zoster.
Face-to-face contact with a
case of chickenpox or
uncovered zoster lesions for at
least 5 minutes.

No ZIG
Oral aciclovir if at risk, ie.
Second half of pregnancy1,2,3
Underlying lung disease4
Immunocompromised5
Smoker6

Advise to seek medical attention immediately if chickenpox develops

Remains
well

Develops
chickenpox

45

ALGORITHM 2

V A R I C E L L A

Z O S T E R

A L G O R I T H M

V I R U S

Management of Varicella Zoster


in Pregnancy
Medical review essential if chickenpox
develops during pregnancy

ALGORITHM 1

No complications*

Complications*
or immunocompromised

< 24 hours since


onset of rash

> 24 hours since


onset of rash

Oral aciclovir

No aciclovir

Low risk group


(see Algorithm 1)

Monitor at home
Advise to seek attention
if complications develop

High risk group


(see Algorithm 1)

Complications
develop

Intravenous aciclovir
Supportive therapy

Complications
develop

Monitor in hospital

RECOVERY

NOTES
* Complications:
Respiratory symptoms
Haemorrhagic rash
Persistent fever >6 days
New pocks developing >6
days
Consensus view + refs 1,7-9
(see Appendix 2 for doses)

Fetal medicine counselling

Consider Caesarean section if:


Fetal compromise
Maternal respiratory failure
exacerbated by advanced
pregnancy

ALGORITHM 3

46

ALGORITHM 4

V A R I C E L L A

Z O S T E R

A L G O R I T H M

V I R U S

Fetal Medicine Counselling


Following Varicella Zoster in Pregnancy
Risk of fetal varicella syndrome (FVS)
following maternal chickenpox in pregnancy

Incidence of FVS following varicella in pregnancy

Large case studies suggest rates of


2-2.8% in first 20 weeks gestation10,11

Timing of maternal infection may be important

< 12 weeks gestation


0.4%

12-20 weeks gestation


2%

>20 weeks gestation Isolated


case reports only

No single diagnostic/prognostic test available


Regular fetal ultrasound for developing anomalies is
recommended12
VZV fetal serology is unhelpful
Consider amniocentesis: ve VZV PCR may be reassuring13

Sequelae of FVS

Frequency10,11

Abnormalities
Skin scars
Eye abnormalities
Limb abnormalities
Prematurity, low birthweight
Cortical atrophy, mental retardation
Poor sphincter control
Early death

47

78%
60%
68%
50%
46%
32%
29%

V A R I C E L L A

Z O S T E R

A L G O R I T H M

V I R U S

Management of Infants from


Mothers with Perinatal Varicella Zoster
Treat infant according to timing
of maternal chickenpox

Maternal chickenpox
> 7 days before delivery

No ZIG required 7
No special interventions
No isolation of infant from
mother
Breast feeding encouraged
Even if baby has chickenpox at
birth or very soon after usually
no interventions are required
unless infant is very preterm*

Maternal chickenpox
7 days before delivery

Very preterm infants born with


chickenpox should receive IV
aciclovir

ZIG required immediately 7,14,15


Should be given < 24 hours
after birth but may be given
up to 72 hours
Discharge term infants as soon
as possible
No other interventions
No isolation of infant from
mother
Breast feeding encouraged

Maternal chickenpox
028 days after delivery

ZIG required immediately 7,14,15


Should be given < 24 hours
after development of maternal
rash but may be given up to
72 hours
Discharge term infants as soon
as possible
No other interventions
No isolation of infant from
mother
Breast feeding encouraged

Develops chickenpoxx

*Very preterm infant


in nursery

NOTES
Transplacentally acquired VZV
is high-risk and severity reduced
by ZIG.14,15
Risk also high for seronegative
baby <28 days old.16,17
ZIG not always effective in
preventing severe disease.16,17
See Appendix 2 for aciclovir
dose.
*Very preterm or = 28 weeks
gestation

Term infant at home


or on postnatal ward

IV aciclovir
ALGORITHM 6

Admit to
paediatric unit

Mild disease and ZIG given


< 24 hours after birth

Keep under observation


Treat with IV aciclovir if
respiratory symptoms develop

48

Severe disease or ZIG given


> 24 hours after birth

Treat with IV aciclovir


Supportive care as required

V A R I C E L L A

Z O S T E R

A L G O R I T H M

V I R U S

Management of Term Neonates Exposed to


Varicella Zoster in the Postnatal Wards or at Home
Is exposure significant?

Same open ward as chickenpox case


1 hour or more close contact with staff
member or visitor with chickenpox or
uncovered herpes zoster lesion

No

No
action
required

Note: chickenpox cases may be infectious in 48 hour period


prior to development of rash

Yes

Has mother had chickenpox previously?

Yes

No or Uncertain

Check maternal serology urgently

Seronegative or
serology unavailable

Seropositive

No intervention required
No isolation from affected sibling required
Review if develops chickenpox

Administer ZIG to infant15,16,17


No isolation from sibling required
Medical review if infant develops
chickenpox
Admit to hospital for intravenous aciclovir
if unwell (tachypnoeic, poor feeding)

NOTES
Exposure to Varicella or Zoster considered to be significant within the neonatal unit or postnatal ward.
Patient sharing the same open ward as case of chickenpox or zoster.
Face-to-face contact with a case of chickenpox or uncovered zoster lesions for at least 5 minutes.
Contact for 1 hour or more with case (either staff or patient) with chickenpox lesions or who developed lesions 24 hours later.

49

V A R I C E L L A

Z O S T E R

A L G O R I T H M

V I R U S

Treatment and Isolation of Infants


Exposed to Varicella Within the Neonatal Unit
Is exposure significant?

Same open ward as varicella case


1 hour or more close contact with staff member or visitor with varicella or
uncovered herpes zoster lesion

No

No
action
required

Note: chickenpox cases may be infectious in 48 hour period prior to development of rash

Yes

28 weeks 18

Administer ZIG

Isolate days 7-28 post exposure


Discharge where possible

>28 weeks

Gestational age at delivery

Urgent maternal serology

Seronegative or
serology not available

Seropositive

ZIG

No ZIG

Isolate days 7-28 post exposure


Discharge where possible

Isolate days 7-21 post exposure


Discharge where possible

Infant develops varicella

ALGORITHM 4 AND 5
for treatment guidelines

Isolate until all


lesions are crusted

NOTES
Immunisation against VZV of susceptible staff strongly recommended.
APPENDIX 1
Varicella zoster immune globulin
High titre varicella zoster immune globulin (ZIG) is available from the
Red Cross Blood Transfusion Service in Australia on a restricted
basis for the prevention of varicella in high risk subjects. Each vial
contains 2 ml (16% solution of gammaglobulin fraction of human
plasma from donors with high titre of varicella antibodies + thiomersal 0.01% w/v). The recommended dose is 2 ml for children 0-5
years, 4 ml for children 6-12 years and 6 ml for adults.7
Administration is by intramuscular injection with few adverse effects
other than local discomfort reported. This can be lessened if the ZIG
is at room temperature when administered. ZIG should never be
given intravenously.7

Ventilated cases
require strict isolation
Consider transfer out of unit
if isolation cubicle not available

APPENDIX 2
Aciclovir
Aciclovir appears to be a safe and relatively well tolerated drug
although it may impair renal function if given to patients who are not
adequately hydrated.9 It is not licensed for use in pregnancy but
appears to be safe9 and its use is indicated in the high risk situations
outlined.
The recommended intravenous dose for treating VZV infection in
adults and infants is 10-20 mg/kg every 8 hours.
The oral dose for adults is 800 mg five times daily. The use of oral
acyclovir in the neonate is not recommended.

50

R E F E R E N C E S
of infants using HBV vaccine in
Shanghai. Preliminary report of a randomized double-blind placebo-controlled trial. Chin. Med. J. Engl. 1985;
98: 6236

Cytomegalovirus

Acknowledgements

References

A/Professor William Rawlinson (SEALS


Pathology, Virology, Prince of Wales
Hospital, Randwick), and Dr Peter
Robertson (SEALS Pathology, Serology,
Prince of Wales Hospital, Randwick) for
comments on diagnosis.

9. http://www.who.int/vaccinesdiseases/diseases/hepatitis_b.htm

Enterovirus

10. Boxall EH, Flewett TH, Dane DS et al.


Letter: Hepatitis-B surface antigen in
breast milk. Lancet 1974; 2: 10078

Rawlinson WD. Broadsheet. Number


50: Diagnosis of human
cytomegalovirus infection and disease.
Pathology 1999; 31(2): 10915.

2. Lazzarotto T, Spezzacatena P, Pradelli


P et al. Avidity of immunoglobulin G
directed against human
cytomegalovirus during primary and
secondary infections in immunocompetent and immunocompromised subjects.
Clin. Diagn. Lab. Immunol. 1997; 4:
46973.

Further Reading
1. Robath, HA. Antiviral therapy for
enteroviral infections. Ped. Infect. Dis.
J. 1999; 18: 6323

3. Donner C, Liesnard C, Brancart F et al.


Accuracy of amniotic fluid testing before
21 weeks gestation in prenatal diagnosis of congenital cytomegalovirus infection. Prenat.Diagn. 1994; 14: 10559.

2. Sawyer MH. Enteroviral Infections. Ped.


Infect. Dis. J. 1999; 18: 103340

4. Lipitz S, Yagel S, Shalev E et al.


Prenatal diagnosis of fetal primary
cytomegalovirus infection. Obstet.
Gynecol. 1997; 89: 7637.

4. Cherry JD. Enteroviruses. In: Infectious


Diseases of the Newborn Infant. 4th
Edn, 40446. Remington JS, Klein JO,
Eds, WB Saunders & Co., Philadelphia,
1995.

5. Gouarin S, Palmer P, Cointe D, et al.


Congenital HCMV infection: a collaborative and comparative study of virus
detection in amniotic fluid by culture and
by PCR. J Clin Virol. 2001; 2: 47-55.

3. Kearns GL et al. Pleconaril


Pharmacokinetics in Neonates Ped.
Infect. Dis. J. 2000; 19: 8338

5. Rotbart HA, Webster D. Treatment of


potentially life-threatening enterovirus
infections with pleconaril. Clin Infect Dis
2001; 32: 228-35

11. Report of the Committee on Infectious


Diseases, 25th ed. Elk Grove, Ill,
American Academy of Pediatrics, p. 300

Acknowledgements
Dr Joe Sasadeusz at Walter and Eliza Hall
Institute, University of Melbourne and Dr
David McIntosh at Wyeth Vaccines,
Maidenhead, UK

Hepatitis C Virus
References
1. Dore GJ, Kaldor JM, McCaughan GW.
Systematic review of role of polymerase
chain reaction in defining infectiousness
among people infected with hepatitis C
virus [see comments]. BMJ. 1997; 315:
3337

6. Lazzarotto T, Varani S, Guerra B et al.


Prenatal indicators of congenital
cytomegalovirus infection. J. Pediatr.
2000; 137:905.

6. Kearns GL, Bradley JS, Jacobs RF et


al. Single dose pharmacokinetics of pleconaril in neonates. Pediatr Infect Dis J
2000; 19: 833-9.

7. Hayes K. Cytomegalovirus: The challenge of a common virus infection.


Med. J. Aust. 1985; 142: 1746.

2. Dore GJ, Pritchard-Jones J, Fisher D et


al.
Who's at risk? Risk factors for HCV
infection in Australia. Aust. Fam.
Physician 1999; 28: S8S13

Hepatitis B Virus

8. Boppana SB, Rivera LB, Fowler KB,


Mach M, Britt WJ. Intrauterine transmission of cytomegalovirus to infants of
women with preconceptional immunity.
N Engl J Med. 2001; 344: 1366-71.

3. Dienstag JL. Sexual and perinatal transmission of hepatitis C. Hepatology


1997; 26: 66S70S

1. Juszczyk J. Clinical course and consequences of hepatitis B infection.


Vaccine 2000; 18 Suppl 1: S23S25

9. Noyola DE, Demmler GJ, Nelson CT, et


al. Early predictors of neurodevelopmental outcome in symptomatic congenital cytomegalovirus infection. J
Pediatr 2001; 138: 325-31.
10. Fowler KB, McCollister FP, Dahle AJ et
al. Progressive and fluctuating sensorineural hearing loss in children with
asymptomatic congenital
cytomegalovirus infection. J. Pediatr.
1997; 130(4): 62430.
11. Ivarsson SA, Lernmark B, Svanberg L.
Ten-year clinical, developmental, and
intellectual follow-up of children with
congenital cytomegalovirus infection
without neurologic symptoms at one
year of age. Pediatrics 1997; 99: 8003.
12. Whitley RJ, Cloud G, Gruber W et al.
Ganciclovir treatment of symptomatic
congenital cytomegalovirus infection:
results of a phase II study. National
Institute of Allergy and Infectious
Diseases Collaborative Antiviral Study
Group. J. Infect. Dis. 1997; 175(5):
10806.

References

2. Schweitzer IL, Dunn AE, Peters RL et


al. Viral hepatitis B in neonates and
infants. Am. J. Med. 1973; 55: 76271
3. Lee SD, Lo KJ, Tsai YT et al. Role of
Caesarean section in prevention of
mother-infant transmission of hepatitis B
virus. Lancet 1988; 2: 8334
4. Towers CV, Asrat T, Rumney P. The
Presence of hepatitis B surface antigen
and deoxyribonucleic acid in amniotic
flueid and cord blood. Am J Obset.
Gyencol. 2001; 184: 1514-8.
5. The Australian Immunisation Handbook,
7th Edn, NH&MRC, Canberra, p. 130
6. Sepkowitz KA. In: Principles and
Practice of Infectious Diseases. 5th
Edn, Mandell, Dougles & Bennett, Eds,
Churchill Livingstone, Philadelphia, p.
3040
7. Theppisai U, Thanuntaseth C, Chiewsilp
P et al. A comparison between the efficacy of passive-active and active immunization for prevention of perinatal
transmission of hepatitis B virus.
J. Med. Assoc. Thai. 1987; 70: 45962.
8. Xu ZY, Francis DP, Liu CB et al.
Prevention of hepatitis B virus carriage

51

4. Spencer JD, Latt N, Beeby PJ et al.


Transmission of hepatitis C virus to
infants of human immunodeficiency
virus-negative intravenous drug-using
mothers: rate of infection and assessment of risk factors for transmission. J.
Viral. Hepat. 1997; 4: 395409.
5. Report of the Committee on Infectious
Diseases, 25th edn. Elk Grove, Ill,
American Academy of Pediatrics, p. 304
6. Polywka S, Schroter M, Feucht HH et
al. Low risk of vertical transmission of
hepatitis C virus by breast milk. Clin.
Infect. Dis. 1999; 29: 13279
7. Kumar RM, Shahul S. Role of breastfeeding in transmission of hepatitis C
virus to infants of HCV-infected mothers. J. Hepatol. 1998; 29: 1917
8. Garland SM, Tabrizi S, Robinson P et
al. Hepatitis C role of perinatal transmission. Aust. NZ J. Obstet. Gynaecol.
1998; 38: 4247

Acknowledgements
Dr Joe Sasadeusz at Walter and Eliza Hall
Institute, University of Melbourne and Dr
David McIntosh at Wyeth Vaccines,
Maidenhead, UK

R E F E R E N C E S
Herpes Simplex Virus
References
1. Arvin AM, Hensleigh PA, Prober CG et
al. Failure of antepartum maternal cultures to predict the infants risk of exposure to herpes simplex virus at delivery.
N Engl. J. Med. 1986; 315: 796800
2. Brown ZA, Benedetti J, Ashley R et al.
Neonatal herpes simplex virus infection
in relation to asymptomatic maternal
infection at the time of labour. N. Engl.
J. Med. 1991; 324: 124752
3. Randolph AG, Washington E, Prober
CG. Cesarian delivery for women presenting with genital herpes lesions.
JAMA, 1993; 270: 7782
4. Brown ZA, Selke A, Zeh J et al. The
acquisition of herpes simplex virus during pregnancy. N. Engl. J. Med 1997;
337: 50915.
5. Nahmias AJ, Josey WE, Naib ZM et al.
Perinatal risk associated with maternal
genital herpes simplex virus infection.
Am. J. Obstet. Gynecol. 1997; 110: 82537.
6. Scott LL, Hollier LM, McIntire D et al.
Acyclovir suppression to prevent clinical
recurrences at delivery after first
episode genital herpes in pregnancy: an
open-label trial. Infect Dis obstet
Gynecol 2001; 9 (2): 75-80.
7. Kimberlin DW, Lin CY, Jacobs RF et al.
Safety and efficacy of high-dose intravenous acyclovir in the management of
neonatal herpes simplex virus infections. Pediatrics. 2001; 108: 230-8.
8. Kimberlin D, Powell D, Gruber W et al.
Administration of oral acyclovir suppressive therapy after neonatal herpes simplex virus disease limited to the skin,
eyes, and mouth: results of a phase I/II
trial. Pediatr. Infect. Dis. J. 1996; 15:
24754
9. Jones CA, Isaacs D, McIntyre P,
Minutillo C, Cunningham A. Neonatal
herpes simplex virus infection. Seventh
Annual Report, Australian Paediatric
Surveillance Unit 1999; 32-3.

Acknowledgements
Associate Professor Suzanne Garland

Human Immunodeficiency
Virus
References
Information pertaining to HIV antiretroviral
management including maternal and perinatal issues is constantly under review.
Updates can be accessed on
http://hivatis.org. Information was last
accessed on February 2002.
1. Ziegler JB. Antenatal screening for HIV
in Australia: time to revise policies? Med
J Aust 1999;171:201-3.
2. Macdonald A, Cruickshank M, Ziegler
JB, Elliot E, Kaldor J. Perinatal expo-

sure to HIV in Australia, 1982-1994.


Med J Australia 1997;166:77-80.
3. Carpenter CC, Cooper DA, Fischl MA,
Gatell JM, Gazzard BG, Hammer SM et
al. Antiretroviral therapy in adults:
updated recommendations of the
International AIDS Society-USA Panel.
JAMA 2000 Jan 2000;283:381-90.
4. Dickover RE, Garraty EM, Herman SA,
Myung-Shin S, Plaeger S, Boyer PJ et
al. Identification of levels of maternal
HIV-1 RNA associated with risk of perinatal transmission Identification of levels of maternal HIV-1 RNA associated
with risk of perinatal transmission.
Effect of maternal zidovudine treatment
on viral load. JAMA 1996;275(8):599605.
5. Garcia PM, Kalish LA, Pitt J, Minkoff H,
Quinn TC, Burchett SK et al. Maternal
levels of plasma human immunodeficiency virus type 1 RNA and the risk of
perinatal transmission. Women and
Infants Transmission Study Group. N
Engl J Med 1999;341:394-402.
6. Newell ML, Dunn DT, Peckham CS,
Semprini AE, Pardi G. Vertical transmission of HIV-1: maternal immune status
and obstetric factors. The European
Collaborative Study. AIDS
1996;10(14):1675-81.

Intrapartum and neonatal single-dose


nevirapine compared with zidovudine
for prevention of mother-to-child transmission of HIV-1 in Kampala, Uganda:
HIVNET 012 randomised trial. Lancet
1999;354:795-802.
13. Connor EM, Sperling RS. Reduction of
maternal-infant transmission of human
immunodeficiency virus type 1 with
zidovudine (AZT) treatment. N Engl J
Med 1994; 331:1173-1180.
14. Jackson JB, Becker-Pergola G, Guay
LA, Musoke P, Mracna M, Fowler MG et
al. Identification of the K103N resistance mutation in Ugandan women
receiving nevirapine to prevent HIV-1
vertical transmission. AIDS
2000;14(11):F111-F115.
15. Dorenbaum, A. Report of results of
PACTG 316: An international Phase III
trial of standard antiretroviral (ARV) prophylaxis plus nevirapine (NVP) for prevention of perinatal HIV transmission.
8th Conference on Retroviruses and
Opportunistic Infections, Chicago .
2001. (Abstract LB7).
16. Palasanthiran P, Robertson P, Ziegler
JB, Graham GG. Decay of transplacental human immunodeficiency virus type
1 antibodies in neonates and infants.
J.Infect.Dis. 1994;170(6):1593-6.

7. Landesman SH, Kalish LA, Burns DN,


Minkoff H, Fox HE, Zorrilla C et al.
Obstetrical factors and the transmission
of human immunodeficiency virus type
1 from mother to child. The Women and
Infants Transmission Study [see comments]. New England Journal of
Medicine 1996;334(25):1617-23.

17. Shaffer N, Chuachoowong R, Mock PA,


Bhadrakom C, Siriwasin W, Young NL
et al. Short-course zidovudine for perinatal HIV-1 transmission in Bangkok,
Thailand: a randomised controlled trial.
Bangkok Collaborative Perinatal HIV
Transmission Study Group. Lancet
1999;353:773-80.

8. The mode of delivery and the risk of


vertical transmission of human immunodeficiency virus type 1 - a meta-analysis of 15 prospective cohort studies.
The International Perinatal HIV Group.
N Engl J Med 1999;340:977-87.

18. Wade NA, Birkhead GS, Warren BL,


Charbonneau TT, French PT, Wang L et
al. Abbreviated regimens of zidovudine
prophylaxis and perinatal transmission
of the human immunodeficiency virus. N
Engl J Med 1998;339:1409-14.

9. Dunn DT, Newell ML, Ades AE,


Peckham CS. Risk of human immunodeficiency virus type 1 transmission
through breastfeeding. Lancet
1992;340(8819):585-8.

Acknowledgements

10. Nduati R, John G, Mbori-Ngacha D,


Richardson B, Overbaugh J, Mwatha A
et al. Effect of breastfeeding and formula feeding on transmission of HIV-1: a
randomized clinical trial. JAMA 2000
Mar 1;283(9):1167-74. 2000;283:116774.
11. Mandelbrot L, Mayaux MJ, Bongain A,
Berrebi A, Moudoub-Jeanpetit Y, Benifla
JL et al. Obstetric factors and motherto-child transmission of human immunodeficiency virus type 1: the French
perinatal cohorts. SEROGEST French
Pediatric HIV Infection Study Group.
American Journal of Obstetrics &
Gynecology 1996;175(3 Pt 1):661-7.
12. Guay LA, Musoke P, Fleming T,
Bagenda D, Allen M, Nakabiito C et al.

52

Associate Professor John Ziegler, Head,


Department of Immunology and Infectious
Diseases, Sydney Childrens Hospital,
Randwick and Dr Dominic Dwyer,
Department of Virology, Westmead Hospital
for perusal and comments.

Listeria
References
1. Gellin BG, Broome CV. Listeriosis.
JAMA 1989; 261: 131320
2. Lorber B. Listeriosis. Clin. Infect. Dis.
1997; 24: 1-11
3. Hof H, Nichterlein T, Kretschmar M.
Management of listeriosis. Clin.
Microbiol. Rev. 1997; 10: 34557
4. Enocksson E, Wretlind B, Sterner G et
al. Listeriosis during pregnancy and in
neonates. Scand. J. Infect. Dis. 1990;
71(Suppl): 8994

R E F E R E N C E S
5. Farmer K, Kreth HW. Listeriosis.
Pediatric Therapy 3rd Edn. Eichenwald
& Stroder, Eds, Mosby 1993; 74951
6. Farmer K. Listeriosis. In: Current therapy in Pediatric Infectious Diseases. 2nd
Edn, Nelson, Ed., Decker 1988; 21012
7. Bortulussi R, Schlech WF. Listeriosis.
In: Infectious Diseases of the Fetus and
Newborn. Remington JS, Klein JO, Eds,
4th Edn, WB Saunders Co.,
Philadelphia. 105573
8. Boucher M, Yonekura ML. Perinatal listeriosis (early-onset): Correlation of
antenatal manifestations and neonatal
outcome. Obstet. Gynecol. 1986; 68:
5937

Mycobacterium Tuberculosis
Further Reading & Sources
1. Hamadeh M, Glassroth J. Tuberculosis
and pregnancy. Chest 1992; 101: 111420
2. Miller K, Miller, J. Tuberculosis in pregnancy: Interactions, diagnosis, and
management. Clin. Obstet. Gynecol.
1996; 39: 12042
3. Brost B, Newman R. The maternal and
fetal effects of tuberculosis therapy.
Obstet. Gynecol. Clin. North Am. 1997;
24: 65973
4. American Academy of Pediatrics
Committee on Infectious Diseases:
Chemotherapy for tuberculosis in
infants and children. Pediatrics 1992;
89: 1615
5. Starke J. Tuberculosis. An old disease
but a new threat to the mother, fetus,
and neonate. Clin. Perinatol. 1997; 24:
10727
6. Chemotherapy and management of
tuberculosis in the United Kingdom:
Recommendations 1998. Joint
Tuberculosis Committee of the British
Thoracic Society. Thorax 1998;.53: 53648
7. Riley L. Pneumonia and tuberculosis in
pregnancy. Infect. Dis. Clin. North Am.
1997; 11: 11933
8. Cantwell MF, Shehab ZM, Costello AM,
et al. Brief Report: congenital tuberculosis. N Engl J Med. 1994; 330: 1051-4
9. Abughali N, Van der Kuyp F, Annable W
et al. Congenital tuberculosis. Pediatr.
Infect. Dis. J. 1994; 13: 73841
10. Voss L. Position paper: Management of
tuberculosis in children. J. Paediatr.
Child Health 2000; 36: 5306

Parvovirus
References
1. Harger JH, Adler SP, Koch WC et al.
Prospective evaluation of 618 pregnant
women exposed to parvovirus B19:

risks and symptoms. Obstet. Gynecol.


1998; 91: 41320
2. Gillespie SM, Cartter ML, Asch S et al.
Occupational risk of human parvovirus
B19 infection for school and day-care
personnel during an outbreak of erythema infectiosum. JAMA 1990; 263:
20615
3. Kelly HA, Siebert D, Hammond R et al.
The age-specific prevalence of human
parvovirus immunity in Victoria,
Australia compared with other parts of
the world. Epidemiol. Infect. 2000; 124:
44957
4. Miller E, Fairley CK, Cohen BJ et al.
Immediate and long term outcome of
human parvovirus B19 infection in pregnancy. Brit. J. Obstet. Gynaecol. 1998;
105: 1748
5. Schild RL, Bald R, Plath H et al.
Intrauterine management of fetal parvovirus B19 infection. Ultrasound
Obstet. Gynecol. 1999; 13: 1616
6. Fairley CK, Smoleniec JS, Caul OE et
al. Observational study of effect of
intrauterine transfusions on outcome of
fetal hydrops after parvovirus B19 infection. Lancet 1995; 346: 13357
7. Rodis JF, Borgida AF, Wilson M et al.
Management of parvovirus infection in
pregnancy and outcomes of hydrops: A
survey of members of the Society of
Perinatal Obstetricians. Am. J. Obstet.
Gynecol. 1998; 179: 9858
8. Koch WC, Harger JH, Barnstein B et
al.. Serologic and virologic evidence for
frequent intrauterine transmission of
human parvovirus B19 with a primary
maternal infection during pregnancy.
Pediatr. Infect. Dis. J. 1998; 17: 48994
9. Cohen BJ, Bates CM. Evaluation of 4
commercial test kits for parvovirus B19specific IgM. J. Virol. Methods 1995; 55:
1125
10. Torok TJ, Wang QY, Gary GWJ et al.
Prenatal diagnosis of intrauterine infection with parvovirus B19 by the polymerase chain reaction technique. Clin.
Infect. Dis. 1992; 14: 14955

Rubella
References
1. Gilbert GL, Field P. What do rubella
antibody tests mean? Inoculum 1997;
6(3): 13
2. Sintchenko V, Field PR, Gilbert GL.
Rubella antibody in antenatal clinic
patients. Aust. NZ J. Med. 1999; 29:
284
3. Morgan-Capner P, Hodgson J,
Hambling, MH et al. Detection of rubella-specific IgM in subclinical rubella
reinfection in pregnancy. Lancet 1985;
1: 2446
4. Cradock-Watson JE, Ridehalgh MKS,
Anderson MJ et al.. Outcome of asymptomatic infection with rubella virus dur-

53

ing pregnancy. J. Hyg. 1981; 87: 14754


5. Miller E, Craddock-Watson JE, Pollock
TM.. Consequences of confirmed
maternal rubella at successive states of
pregnancy. Lancet 1982; ii: 7814
6. Enders G, Nickerl-Pacher U, Miller E et
al. Outcome of confirmed periconceptional maternal rubella. Lancet 1988; i:
14457
7. Peckham C. Clinical and laboratory
study of children exposed in utero to
maternal rubella. Arch Dis Child 1972;
47: 5717
8. Infectious disease in pregnancy and the
newborn infant. Gilbert, GL, Chur,
Harvard Academic Publishers,
Switzerland, New York, USA. 1990
9. Infectious Disease of the Fetus and
Newborn, 5th Edn, Remington J, WB
Saunders, 2000
10. Daffos F, Forestier F, Grangeot-Kerros L
et al. Prenatal diagnosis of congenital
rubella. Lancet 1984: 2: 13
11. Revello MG, Baldanti F, Sarasini A et al.
Prenatal diagnosis of rubella virus infection by direct detection and semiquantitation of viral RNA in clinical samples by
reverse transcription. J. Clin. Microbiol.
1997; 35(3): 70813
12. Enders G, Jonatha W. Prenatal diagnosis of intrauterine rubella. Infection;
1987; 15: 1624
13. Menser MA, Forrest JM. Rubella- high
incidence of defects in children considered normal at birth. Med. J. Aust.
1974; 1: 1236

Acknowledgements
Professor Lyn Gilbert and Professor
Margaret Burgess, Westmead Hospital,
Westmead NSW

Streptococcus, Group B
References and Sources
1. Prevention of perinatal group B streptococcal disease: a public health perspective. Centers for Disease Control and
Prevention. MMWR Morb Mortal Wkly
Rep 1996; 45(RR-7): 1-24
2. Boyer KM, Gotoff SP. Prevention of
early-onset neonatal group B streptococcal disease with selective intrapartum chemoprophylaxis. N. Engl. J.
Med. 1986; 314(26): 16659
3. Schrag SJ, Zywicki S, Farley MM et al.
Group B streptococcal disease in the
era of intrapartum antibiotic prophylaxis.
N. Engl. J. Med. 2000; 342: 1520
4. Centers for Disease Control and
Prevention. 2002. Prevention of perinatal group B streptococcal disease:
Revised guidelines from the Centers for
Disease Control and Prevention.
MMWR Recommendations and
Reports: In press.

R E F E R E N C E S
5. Yancey MK, Schuchat A, Brown LK et al.
The accuracy of late antenatal screening
cultures in predicting genital group B
streptococcal colonization at delivery.
Obstet. Gynecol. 1996; 88: 8115.
6. Boyer KM, Gadzala CA, Kelly PD et al.
Selective intrapartum chemoprophylaxis
of neonatal group B streptococcal earlyonset disease. II. Predictive value of
prenatal cultures. J. Infect. Dis. 1983;
148: 8029
7. Garland SM, Kelly N, Ugoni AM. Is
antenatal group B streptococcal carriage a predictor of adverse obstetric
outcome? Infect. Dis. Obstet. Gynecol.
2000; 8: 13842
8. Rosa-Fraile M, Rodriguez-Granger J,
Cueto-Lopez M et al. Use of Granada
medium to detect group B streptococcal
colonization in pregnant women. J. Clin.
Microbiol. 1999; 37: 267477
9. Schuchat A, Deaver-Robinson K,
Plikaytis BD et al. Multistate case-control study of maternal risk factors for
neonatal group B streptococcal disease.
The Active Surveillance Study Group.
Pediatr. Infect. Dis. J. 1994; 13: 6239
10. Boyer KM, Gadzala CA, Kelly PD, et al.
Selective intrapartum chemoprophylaxis
of neonatal group B streptococcal earlyonset disease. III. Interruption of motherto-infant transmission. J Infect Dis.
1983; 148: 810-6

Toxoplasma
References
1. Dunn D, Wallon M, Peyron F et al.
Mother-to-child transmission of toxoplasmosis: Risk estimates for clinical counselling. Lancet 1999; 353: 182933
2. Foulon W, Pinon JM, Stray-Pedersen B
et al. Prenatal diagnosis of congenital
toxoplasmosis: a multicenter evaluation
of different diagnostic parameters. Am.
J. Obstet. Gynecol. 1999b; 181: 8437
3. Robert-Gangneux F, Gavinet MF,
Ancelle T et al. Value of prenatal diagnosis and early postnatal diagnosis of congenital toxoplasmosis: retrospective
study of 110 cases. J. Clin. Microbiol.
1999; 37: 28938
4. Naessens A, Jenum PA, Pollak A et al.
Diagnosis of congenital toxoplasmosis in
the neonatal period: A multicenter evaluation. J. Pediatr. 1999;135: 7149
5. Foulon W, Villena I, Stray-Pedersen B et
al. Treatment of toxoplasmosis during
pregnancy: A multicenter study of impact
on fetal transmission and childrens
sequelae at age 1 year. Am. J. Obstet.
Gynecol. 1999a; 180: 4105
6. Wallon M, Liou C, Garner P et al.
Congenital toxoplasmosis: systematic
review of evidence of efficacy of treatment in pregnancy. BMJ 1999; 318:
15114
7. Petersen E, Eaton RB. Control of con-

genital infection with Toxoplasma gondii


by neonatal screening based on detection of specific immunoglobulin M antibodies eluted from phenylketonuria filterpaper blood-spot samples. Acta
Paediatr. Suppl. 1999; 88: 369

Treponema Pallidum
(Syphilis)
Further Reading
Centers for Disease Control and Prevention.
1998 Guidelines for treatment of sexually
transmitted diseases. MMWR 1998; 47(No.
RR-1): 2849
Hollier LM, Cox SM. Syphilis. Semin.
Perinatol. 1998; 22: 32331
Humphrey MD, Bradford DL. Congenital
syphilis: Still a reality in 1996. Med. J. Aust.
1996; 165: 3825
Larsen SA, Steiner BM, Rudolph AH.
Laboratory diagnosis and interpretation of
tests for syphilis. Clin. Microbiol. Rev. 1995;
8; 121
Romanowski B, Sutherland R, Fick et al.
Serologic response to treatment of infectious
syphilis. Ann. Intern. Med. 1991; 114: 10059
Sanches PJ. Syphilis and Pregnancy in
Sexually Transmitted Diseases and Adverse
Outcomes of Pregnancy. ASM Press,
Washington 1999; 12550
Sanchez PJ, Wendel GD, Grimprel E et al.
Evaluation of molecular methodologies and
rabbit infectivity testing for the diagnosis of
congenital syphilis and neonatal central
nervous system invasion by Treponema
pallidum. J. Infect. Dis. 1993; 167: 14857
Sheffield JS, Wendel GD. Syphilis in pregnancy. Clin. Obstet. Gynecol. 1999; 42: 97106
Stoll BJ, Francis KL, Larsen S et al. Clinical
and serological evaluation of neonates for
congenital yphilis: a continuing diagnostic
dilemma. J. Infect. Dis. 1993; 167: 10939
Therapeutic Guidelines: Antibiotic.
Therapeutic Guidelines Ltd, Melbourne, 11th
Edn 2000: 73-4.

Varicella Zoster Virus

fatal chickenpox during prednisolone


treatment. Br. Med. J. 1994; 309: 106970
6. Grayson ML, Newton-John H. Smoking
and varicella pneumonia. .J Infect. 1988;
16: 312
7. The Australian Immunisation handbook.
7th Edn. Canberra, Australian
Government Publishing Service, 2000
8. Lin TY, Huang YC, Ning HC et al. Oral
aciclovir prophylaxis after intimate contact. Pediatr. Infect. Dis. J. 1997; 16:
11625
9. Andrews EB, Yankasksas BC, Cordero
JF et al. Aciclovir in pregnancy registry:
6 years experience. The aciclovir in
pregnancy registry advisory committee.
Obstet. Gynecol. 1991; 78: 11126
10. Pastuszak AL, Levy M, Schick B et al.
Outcome after maternal varicella infection in the first 20 weeks of pregnancy.
N. Engl. J. Med.1994; 330: 9015
11. Jones KL, Johnson KA, Chambers CD.
Offspring of women infected with varicella during pregnancy: a prospective
study. Teratology 1994; 49: 2932
12. Pretorius DH, Hayward I, Jones KL et al.
Sonographic evaluation of pregnancies
with maternal varicella infection. J.
Ultrasound Med. 1992; 11: 45963
13. Mouly F, Mirlesse V, Meritet J et al.
Prenatal diagnosis of fetal varicella
zoster virus infection with polymerase
chain reaction of amniotic fluid in 107
cases. Am. J. Obstet. Gynecol. 1997;
177: 8948
14. Miller E, Cradock-Watson JE et al.
Outcome of newborn babies given anti
varicella zoster immunoglobulin after
peronatal maternal infection with varicella zoster virus. Lancet 1989; ii: 3713
15. Hanngren K, Grandien M, Granstrom G.
Effect of zoster immunoglobulin for varicella prophylaxis in the newborn. Scand.
J. Infect Dis. 1985; 17: 3437
16. Rubin L. Disseminated varicella in the
neonate and implications for immunoprophylaxis in neonates exposed to varicella. Pediatr. Infect. Dis. J. 1986; 56:
1002

1. Gilbert GL. Chickenpox during pregnancy. BMJ 1993; 306: 1079-80

17. Reynolds L, Struik S, Nadel S. Neonatal


varicella: varicella zoster immunoglobulin (VZIG) does not prevent disease.
Arch. Dis. Child Fetal Neonatal Ed.
1999; 81: F69F70

2. Preblud SR, Cochi SL, Orenstein WA.


Varicella zoster infection in pregnancy.
N. Engl. J. Med. 1986; 315: 14167

18. Conway SP, Dear PRF, Smith I.


Immunoglobulin profile of the preterm
baby. Arch. Dis. Child 1985; 60: 20812

References

3. Nathwani D, Maclean A, Conway S,


Carrington D. Varicella infectiouns in
pregnancy and the newborn. J Infect
1998; 36 (supp 1): 59-71.
4. Haake DA, Zakowski PC, Haake DL et al.
Early treatment with acyclovir for varicella
pneumonia in otherwise healthy adults.
Rev. Infect. Dis. 1990; 12: 78897
5. Rice P, Simmons K, Carr R et al. Near

54

Emendations: References
E1. Fowler KB, Stagno S, Pass RF. Maternal immunity and prevention of congenital
cytomegalovirus infection. JAMA.. 2003;289:1008-11
E2. Fowler KB, Stagno S, Pass RF. Interval between births and risk of congenital cytomegalovirus
infection. Clinical Infectious Diseases. 2004;38:1035-7
E3. Kimberlin DW, Lin CY, Sanchez PJ, Demmler GJ, Dankner W, Shelton M et al. Effect of
ganciclovir therapy on hearing in symptomatic congenital cytomegalovirus disease involving the
central nervous system: a randomized, controlled trial. Journal of Pediatrics. 2003;143:16-25
E4. Abzug MJ, Cloud G, Bradley J, Sanchez PJ, Romero J, Powell D et al. Double blind placebocontrolled trial of pleconaril in infants with enterovirus meningitis. Pediatric Infectious Disease
Journal. 2003;22:335-41
E5.

Chapter 3.9 Hepatitis B. The Australian Immunisation Handbook. 8th edition, 2003.

E6. Hardikar W, Elliott EJ, Jones CA. The silent infection: should we be testing for perinatal
hepatitis C and, if so, how? Med.J.Aust. 2006;184: 54-5
E7. Dunn DT, Gibb DM, Healy M, Goodall RL, Butler K, Cafferkey M et al. Timing and
interpretation of tests for diagnosing perinatally acquired hepatitis C virus infection. Pediatric
Infectious Disease Journal. 2001;20:715-6
E8. Polywka S, Pembrey L, Tovo PA, Newell ML. Accuracy of HCV-RNA PCR tests for diagnosis
or exclusion of vertically acquired HCV infection. J.Med.Virol. 2006;78:305-10
E9. Brown ZA, Wald A, Morrow RA, Selke S, Zeh J, Corey L. Effect of serologic status and
cesarean delivery on transmission rates of herpes simplex virus from mother to infant JAMA.
2003;289:203-9
E10. Lyons F, Hopkins S, Kelleher B, McGeary A, Sheehan G, Geoghegan J et al. Maternal
hepatotoxicity with nevirapine as part of combination antiretroviral therapy in pregnancy.
HIV.Medicine. 2006;7:255-60

cover

29/8/05

5:21 PM

Page 1

An infection during pregnancy that could


damage the fetus or infant is much less
common than suspicion or fear of infection.
Vague, nonspecific symptoms malaise,
aches and pains, headache, tiredness, nausea
- are the hallmarks of many vertically
transmissible maternal infections but they are
also common during pregnancy or in any
busy, stressful life. In pregnant women they
cant be dismissed as trivial as they may be in
others.
This publication represents a consensus
among specialists in perinatal infection. It has
been endorsed by the Australasian Society
for Infectious Diseases and the Royal College
of Obstetricians and Gynaecologists of
Australia and New Zealand. We hope it will
not discourage nonspecialists from referring
pregnant women with suspected infection
during pregnancy, but that it will help to guide
the initial assessment and investigation of the
patient, and prevent unnecessary anxiety or
hasty decisions.

Management of
Perinatal Infections
Cytomegalovirus
Enterovirus
Hepatitis B
Hepatitis C
Herpes Simplex Virus
Human Immunodeficiency Virus
Listeria
Mycobacterium Tuberculosis
Parvovirus
Rubella
Streptococcus - Group B
Toxoplasma
Treponema Pallidum (Syphilis)
Varicella Zoster Virus

Edited by Dr Pamela Palasanthiran,


Dr Mike Starr, and Dr Cheryl Jones
Introduction by Prof Lyn Gilbert
AUSTRALASIAN SOCIETY FOR
INFECTIOUS DISEASES 2002

AUSTRALASIAN SOCIETY FOR


INFECTIOUS DISEASES 2002

You might also like