You are on page 1of 17

CLINICAL REPORT Guidance for the Clinician in Rendering Pediatric Care

Management of Infants at Risk for


Group B Streptococcal Disease
Karen M. Puopolo, MD, PhD, FAAP,a,b Ruth Lynfield, MD, FAAP,c James J. Cummings, MD, MS, FAAP,d COMMITTEE ON FETUS AND
NEWBORN, COMMITTEE ON INFECTIOUS DISEASES

Group B streptococcal (GBS) infection remains the most common cause of abstract
neonatal early-onset sepsis and a significant cause of late-onset sepsis among
young infants. Administration of intrapartum antibiotic prophylaxis is the only
a
currently available effective strategy for the prevention of perinatal GBS early- Department of Pediatrics, Perelman School of Medicine, University of
Pennsylvania, Philadelphia, Pennsylvania; bChildren’s Hospital of
onset disease, and there is no effective approach for the prevention of late-onset Philadelphia, Philadelphia, Pennsylvania; cMinnesota Department of
disease. The American Academy of Pediatrics joins with the American College of Health, St Paul, Minnesota; and dDepartments of Pediatrics and
Bioethics, Alden March Bioethics Institute, Albany Medical College,
Obstetricians and Gynecologists to reaffirm the use of universal antenatal Albany, New York
microbiologic-based testing for the detection of maternal GBS colonization to
This document is copyrighted and is property of the American
facilitate appropriate administration of intrapartum antibiotic prophylaxis. The Academy of Pediatrics and its Board of Directors. All authors have filed
purpose of this clinical report is to provide neonatal clinicians with updated conflict of interest statements with the American Academy of
Pediatrics. Any conflicts have been resolved through a process
information regarding the epidemiology of GBS disease as well current approved by the Board of Directors. The American Academy of
Pediatrics has neither solicited nor accepted any commercial
recommendations for the evaluation of newborn infants at risk for GBS disease involvement in the development of the content of this publication.
and for treatment of those with confirmed GBS infection. This clinical report is
Clinical reports from the American Academy of Pediatrics benefit from
endorsed by the American College of Obstetricians and Gynecologists (ACOG), expertise and resources of liaisons and internal (AAP) and external
reviewers. However, clinical reports from the American Academy of
July 2019, and should be construed as ACOG clinical guidance. Pediatrics may not reflect the views of the liaisons or the
organizations or government agencies that they represent.

The guidance in this report does not indicate an exclusive course of


treatment or serve as a standard of medical care. Variations, taking
The Centers for Disease Control and Prevention (CDC) first published into account individual circumstances, may be appropriate.

consensus guidelines on the prevention of perinatal group B streptococcal All clinical reports from the American Academy of Pediatrics
automatically expire 5 years after publication unless reaffirmed,
(GBS) disease in 1996. These guidelines were developed in collaboration revised, or retired at or before that time.
with the American Academy of Pediatrics (AAP), the American College of
DOI: https://doi.org/10.1542/peds.2019-1881
Obstetricians and Gynecologists (ACOG), the American College of Nurse-
Midwives, the American Academy of Family Physicians, and other Address correspondence to Karen M. Puopolo, MD, PhD. E-mail:
puopolok@email.chop.edu
stakeholder organizations1 on the basis of available evidence as well as
PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online, 1098-4275).
expert opinion. The 1996 consensus guidelines recommended either an
antenatal culture–based or risk factor–based approach for the Copyright © 2019 by the American Academy of Pediatrics
administration of intrapartum antibiotic prophylaxis (IAP) to prevent
invasive neonatal GBS early-onset disease (EOD).1 The guidelines were To cite: Puopolo KM, Lynfield R, Cummings JJ, AAP
updated in 2002 primarily on the basis of new data from a CDC multistate COMMITTEE ON FETUS AND NEWBORN, AAP COMMITTEE ON
retrospective cohort study in which authors found universal screening for INFECTIOUS DISEASES. Management of Infants at Risk for
Group B Streptococcal Disease. Pediatrics. 2019;144(2):
group B streptococci (Streptococcus agalactiae) was .50% more effective
e20191881
at preventing the disease compared to a risk-based approach.2,3 In 2010,

PEDIATRICS Volume 144, number 2, August 2019:e20191881 FROM THE AMERICAN ACADEMY OF PEDIATRICS
Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
the CDC once again revised the GBS should be construed as ACOG clinical cases per 100 000 live births in the
perinatal prevention guidelines and guidance. ABCs. Although potentially an
continued to endorse the universal underestimate, this finding was
antenatal culture–based approach to consistent with data from the
identify women who would receive CURRENT EPIDEMIOLOGY OF NEONATAL Northern California Kaiser
GBS INFECTION
IAP to prevent GBS EOD.4 Notable Permanente health care system,
changes in the 2010 revision Early-Onset GBS Infection which found the incidence of
addressed the use of IAP for women newborn infants readmitted to the
GBS EOD is defined as isolation of
in preterm labor and those with hospital within the first week after
group B Streptococcus organisms
preterm prelabor rupture of hospital discharge with culture-
from blood, cerebrospinal fluid (CSF),
membranes (PROM), the choice of confirmed infection attributable to
or another normally sterile site from
specific antibiotics for IAP, and the any bacteria to be approximately 5
birth through 6 days of age.8 Active
use of nucleic acid amplification tests cases per 100 000 live births.14
Bacterial Core surveillance (ABCs),
(NAATs) to identify maternal GBS Globally, outside the United States, an
performed in collaboration with the
colonization. The 2010 revision estimated 200 000 cases of GBS EOD
CDC in 10 states from 2006 to 2015,
included a neonatal management occurred in 2015. Stillbirth, GBS EOD,
found that the overall incidence of
algorithm for secondary prevention of and late-onset GBS cases combined
GBS EOD declined from 0.37 cases
GBS EOD that was widely adopted by contribute to an estimated 150 000
per 1000 live births in 2006 to 0.23
neonatal physicians as a means of fetal and neonatal deaths throughout
cases per 1000 live births in 2015.6
managing risk of all bacterial causes the world, with the largest
Meningitis was diagnosed in 9.5% of
of early-onset sepsis (EOS).5 With concentration of GBS perinatal deaths
infants with GBS EOD. CSF culture-
implementation of universal maternal occurring in Africa.15
positive GBS EOD occurred in the
antenatal screening and IAP, the
absence of bacteremia in 9.1% of Late-Onset GBS Disease
national incidence of GBS EOD has
early-onset meningitis cases
declined from 1.8 cases per 1000 live GBS late-onset disease (LOD) is
(incidence: approximately 2.5 cases
births in 1990 to 0.23 cases per 1000 defined as isolation of GBS from
per 1 million live births).6 Infants
live births in 2015.6 a normally sterile site from 7 to
born at ,37 weeks’ gestation account
89 days of age.8 Rarely, very–late-
Evolving epidemiology, newly for 28% of all GBS cases;
onset GBS disease may occur after
published data, and changing practice approximately 15% of cases occur
3 months of age, primarily among
standards inform periodic review among preterm infants with very low
infants born very preterm or infants
of practice guidelines. In 2017, birth weight (,1500 g).6,9 Overall,
with immunodeficiency syndromes.16,
representatives from the CDC, AAP, GBS infection accounts for 17
GBS LOD rates have not changed
ACOG, and other stakeholder approximately 45% of all cases of
with widespread use of IAP. GBS LOD
organizations agreed to review and culture-confirmed EOS among term
incidence was stable over the
revise the 2010 GBS guidelines. A infants and approximately 25% of all
2006–2015 ABCs study period, with
consensus was reached that the EOS cases that occur among infants
an average incidence of 0.31 cases
AAP would revise neonatal care with very low birth weight.9,10 Death
per 1000 live births.6 The median age
recommendations and ACOG would attributable to GBS EOD occurs
at presentation with GBS LOD was
revise obstetric care guidelines. These primarily among preterm infants: the
34 days (interquartile range: 20–49
separate but aligned publications current case fatality ratio is 2.1%
days). Bacteremia was identified in
replace the CDC 2010 GBS perinatal among term infants and 19.2%
approximately 93% of GBS LOD, and
guidance. among those born at ,37 weeks’
bacteremia without focus was the
gestation.6
This clinical report addresses the most common form of disease. Group
epidemiology, microbiology, disease GBS EOD primarily presents clinically B streptococci were isolated from CSF
pathogenesis, and management at or shortly after birth. The majority in 20.7% of cases, and meningitis was
strategies for neonatal early- and late- of infants become symptomatic by 12 diagnosed in 31.4% of cases. Cultures
onset GBS infection. Maternal to 24 hours of age11–13; during the of bone and joint and peritoneal fluid
management is addressed in ACOG 2006–2015 period of ABCs, 94.7% of yielded group B streptococci in
Committee Opinion No. 782, GBS EOD cases were diagnosed #48 1.8% of cases. CSF culture-positive
“Prevention of Group B Streptococcal hours after birth.6 The incidence of GBS LOD occurred in the absence
Early-Onset Disease in Newborns.”7 newborn infants discharged from of bacteremia in approximately
This clinical report is endorsed by the a birth hospital and readmitted to 20% of late-onset meningitis cases
American College of Obstetricians and a hospital with GBS EOD within (incidence: 1.9 cases per 100 000 live
Gynecologists (ACOG), July 2019, and 6 days of age was approximately 0.3 births). Infants born at ,37 weeks’

2 FROM THE AMERICAN ACADEMY OF PEDIATRICS


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
gestation account for approximately pregnant and nonpregnant women.28, vaginal examinations, invasive fetal
29
42% of all GBS LOD cases, and death Transmission of group B monitoring, and membrane sweeping,
attributable to GBS LOD occurs in streptococci from mother to infant have been associated with GBS EOD in
preterm infants at roughly twice the usually occurs shortly before or some observational studies.39,44 Such
rate of term infants (7.8% vs 3.4%, during delivery. In the absence of IAP, studies are difficult to interpret
respectively).6 GBS LOD complicated approximately 50% of newborn because of confounding; available data
by meningitis has a higher patient infants born to mothers positive for are not sufficient to determine if these
fatality rate than those with other GBS become colonized with group B procedures are associated with an
syndromes. streptococci, and of those, 1% to 2% increased risk for GBS EOD. Current
will develop GBS EOD.30–33 ACOG guidance addresses the
appropriate use of these procedures
PATHOGENESIS OF AND RISK FACTORS Multiple clinical characteristics among GBS-colonized women.7
FOR GBS INFECTION associated with greater risk of Administration of IAP in women with
EOD maternal GBS colonization and with GBS colonization minimizes the impact
the pathogenesis of GBS EOD are of intrapartum obstetric procedures on
Group B Streptococcus emerged as the
predictive of neonatal disease.20,21,34–43 the risk of neonatal GBS EOD.
primary bacterial cause of EOS in the
Lower gestational age is associated
1970s, and subsequent studies LOD
with less effective opsonic, neutrophil-
identified maternal GBS colonization
mediated defenses in the infant as well A positive GBS screen result in the
as the primary risk factor for GBS-
as lower levels of protective maternally mother at the time of birth and at the
specific EOS.18–20 The most common
derived antibody.44–46 Increased time of LOD diagnosis is significantly
pathogenesis of GBS EOD is that of
duration of rupture of membranes associated with LOD.48,49 Maternal
ascending colonization of the uterine
(ROM) promotes the process of colonization is not universally present
compartment with group B
ascending colonization and infection of in LOD cases, however, suggesting that
streptococci that are present in the
the uterine compartment and fetus. horizontal acquisition of group B
maternal gastrointestinal and
Maternal intrapartum fever may reflect streptococci from nonmaternal
genitourinary flora. Infection occurs
the maternal inflammatory response to caregivers may also be part of the
with subsequent colonization and
evolving intraamniotic bacterial pathogenesis of GBS LOD. GBS LOD is
invasive infection of the fetus and/or
infection and is an important predictor strongly associated with preterm
fetal aspiration of infected amniotic
of neonatal early-onset infection. birth.6,48–53 In studies from
fluid. This pathogenesis primarily
African American race and, less Washington in 1992 to 2011 and
occurs during labor for term infants,
consistently, a maternal age ,20 years Houston in 1995 to 2000, it was found
but the timing is less certain among
have been associated with a higher risk that the risk for LOD increased for
preterm infants for whom
of GBS EOD.10,34,37,39 The independent each week of decreasing gestation and
intraamniotic infection may be the
contribution of these factors remains 40% to 50% of all LOD occurs among
cause of PROM and/or preterm
unclear because maternal age and race infants born ,37 weeks’ gestation.48,
labor.21 Rarely, GBS EOD may develop
are also associated with higher rates of 50
Maternal age ,20 years and African
at or near term before the onset of
GBS colonization, preterm birth, and American race are variably associated
labor, potentially because of group B
socioeconomic disadvantage, and with neonatal GBS LOD in United
streptococci traversing exposed but
African American race has been States studies.10,48,50,51 Other clinical
intact membranes. GBS EOD also is
associated with a greater likelihood of intrapartum factors predictive of GBS
associated with stillbirth.22,23
missed antenatal screening.37,41 The EOD (maternal intrapartum fever,
Maternal colonization is delivery of a previous infant with GBS duration of ROM) are not predictive of
a prerequisite for GBS EOD. Authors EOD is associated with increased risk LOD. Authors of a population-based
of a recent meta-analysis estimated in a subsequent delivery,3 a factor that study in Italy found that group B
that globally, GBS colonization was may be related to poor maternal streptococci could be cultured from
detected at vaginal and/or rectal sites antibody responses to colonizing a mother’s milk in approximately 25%
in 18% of pregnant women, with strains or other immune- or strain- of cases that occurred in breastfed
regional variation of 11% to 35%.24 specific factors.42 GBS bacteriuria is infants,49 and authors of case reports
Authors of most clinical studies in the associated with a high level of maternal associate colonized human milk with
United States have found colonization colonization and increased risk of GBS LOD.54 However, human
rates of 20% to 30% among pregnant neonatal colonization and disease.41,47 milk–associated GBS antibody is
women,14,25–28 with variation by Finally, obstetric practices that may protective against GBS LOD,55 and it
maternal age and race. Colonization promote ascending bacterial infection, remains unclear whether human milk
can be ongoing or intermittent among such as the frequency of intrapartum is simply a marker of heavy maternal

PEDIATRICS Volume 144, number 2, August 2019 3


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
and infant colonization or a source of IAP FOR THE PREVENTION OF determine maternal colonization
infection. EARLY-ONSET GBS INFECTION status, both because of variable
reported sensitivity of the point-of-
GBS Virulence Prevention of Perinatal GBS Infection
care NAAT as compared to
Bacterial factors promote invasive Multiple observational studies and 1 traditional culture and because most
GBS infection. Group B streptococci randomized controlled trial have NAAT-based testing cannot be used
are characterized by immunologically revealed that the administration of to determine the antibiotic
distinct surface polysaccharide intrapartum antibiotics before susceptibility of colonizing GBS
capsules that define 10 serotypes delivery interrupts vertical isolates among women with
(types I, Ia, and II–IX). Worldwide, transmission of group B streptococci a penicillin allergy.
serotypes I–V account for 98% of and decreases the incidence of
carriage and 97% of infant invasive invasive GBS EOD.30–32,63 IAP is Updated ACOG recommendations
strains; serotype III accounts for hypothesized to prevent neonatal GBS address the indications for IAP.7 IAP at
approximately 25% of colonizing disease in 3 ways: (1) by temporarily the time of presentation for delivery is
strains and approximately 62% of decreasing maternal vaginal GBS indicated for all women with GBS
invasive infant strains, with regional colonization burden; (2) by preventing colonization identified by antenatal
variation.24,56 Surveillance data in the surface and mucus membrane vaginal-rectal culture, for women with
United States for invasive strains colonization of the fetus or newborn; GBS bacteriuria identified at any point
from 2006 to 2015 revealed that and (3) by reaching levels in newborn during pregnancy, for women with
93.1% of GBS EOD cases were bloodstream above the minimum a history of a previous infant with GBS
attributable to serotypes Ia (27.3%), inhibitory concentration (MIC) of the disease, and for women who present
III (27.3%), II (15.6%), V (14.2%), antibiotic for killing group B in preterm labor and/or with PROM at
and Ib (8.8%); the proportion streptococci.30,31 Current clinical ,37 0/7 weeks’ gestation. Women
attributable to the emerging serotype practices are focused on the who present in labor at $37 0/7
IV ranged from 3.4% to 11.3% over identification of women at highest risk weeks’ gestation with unknown GBS
the study period.6 Serotype III of GBS colonization and/or of status should receive IAP if risk factors
accounted for approximately 56.2% transmission of group B streptococci develop during labor (maternal
of 1387 GBS LOD cases during to the newborn infant to facilitate intrapartum temperature $100.4°F
2006–2015, with serotypes Ia (20%), targeted administration of IAP. [38°C] or duration of ROM $18 hours)
V (8.3%), IV (6.2%), and Ib (6.1%) or if the result of an available point-of-
making up most of the remaining The ACOG currently recommends care NAAT is positive for group B
serotypes. The capsular universal antenatal testing of streptococci. If a woman with
polysaccharide of all GBS serotypes pregnant women for GBS colonization unknown status has a negative point-
resists complement deposition and by using vaginal-rectal cultures of-care NAAT test result but develops
inhibits opsonophagocytosis. obtained at 36 0/7 to 37 6/7 weeks’ intrapartum risk factors, IAP should be
Maternally derived, serotype-specific gestation.7 GBS testing is also administered because the sensitivity of
antibody to maternal colonizing GBS recommended for pregnant women the NAAT may be decreased without
isolates is protective against newborn who present in preterm labor and/or an enrichment incubation step.
infection.44,45 Group B streptococci with PROM before 37 0/7 weeks’ Women with GBS colonization in one
express multiple additional virulence gestation. If maternal GBS pregnancy have an estimated 50% risk
factors, including surface proteins colonization is identified by antenatal of colonization in a subsequent
such as the a and b C-proteins that urine culture, it does not need to be pregnancy.64 Therefore, current ACOG
promote adherence and immune reconfirmed by vaginal-rectal culture. recommendations state that if
evasion, pore-forming toxins such as GBS vaginal-rectal culture is a woman with unknown GBS status
b-hemolysin and CAMP factor, and optimally performed by using a broth presents in labor and is known to have
secreted proteases such as the C5a enrichment step, followed by GBS had GBS colonization in a previous
peptidase that cleaves complement.57 identification by using traditional pregnancy, IAP may be considered.
Strains vary in their expression of microbiologic methods or by NAAT-
virulence factors, many of which are based methods. At some centers, Recommended Antibiotics for GBS
highly regulated by 2-component NAATs may be used to perform real- IAP to Prevent Neonatal GBS EOD
regulatory systems.58–62 The time, point-of-care screening of Group B streptococci remain
hypervirulent serotype III multilocus women who present in labor with susceptible to b-lactam antibiotics,
sequence type 17 (ST17), for unknown GBS status. Point-of care and penicillin G and ampicillin are the
example, is commonly found in cases NAAT-based screening is not the antibiotics best studied for
of GBS meningitis.6,23 primary recommended approach to prevention of neonatal infection.

4 FROM THE AMERICAN ACADEMY OF PEDIATRICS


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
Penicillin G administered to the resistance, erythromycin is no longer maternal and cord blood vancomycin
mother readily crosses the placenta, recommended for GBS IAP.7 concentrations were achieved in most
reaching peak cord blood Antibiotic susceptibility data for the cases with weight-based dosing. The
concentrations by 1 hour and rapidly maternal colonizing GBS isolate ACOG currently recommends weight-
declining by 4 hours, reflecting should be available to support the use based dosing for vancomycin when
elimination of the antibiotic by the of clindamycin for IAP in women who this agent is indicated for IAP.
fetal kidney into amniotic fluid.65 report a penicillin allergy. Several
Current data support the use of
Ampicillin has been detected in cord studies inform the potential efficacy
clindamycin and vancomycin as
blood within 30 minutes and in of clindamycin prophylaxis.72–75
alternative medications for GBS IAP
amniotic fluid within 45 minutes of Authors of 1 study of 21 women
when maternal allergy precludes the
administration to the mother.30 colonized with clindamycin-susceptible
use of b-lactam antibiotics. These
Ampicillin concentrations measured GBS isolates found vaginal GBS colony
medications are likely to provide
in 115 newborn infants at 4 hours of counts declined with administration of
some protection against GBS infection
age were found to be greater than the clindamycin IAP.72 Clindamycin
for both mother and newborn infant
GBS MIC if maternal IAP dosing administered intravenously to 23
when antimicrobial susceptibility
occurred at least 15 minutes before women for high-risk penicillin allergy
testing supports the use of these
delivery.66 Ampicillin IAP decreases resulted in cord blood concentrations
second-line agents. In addition, new
maternal vaginal colonization and that were 37% to 160% of maternal
ACOG recommendations encouraging
prevents neonatal surface concentrations and within therapeutic
the use of penicillin allergy testing in
colonization in 97% of cases if IAP is ranges in 22 of the 23.75 However,
pregnant women with uncertain or
administered at least 2 hours before clindamycin undergoes hepatic
undocumented histories of penicillin
delivery.30,32 Penicillin G has metabolism and is poorly excreted in
reactions are intended to increase the
a narrower antimicrobial spectrum fetal urine and, therefore, does not
number of pregnant women who can
compared to ampicillin and therefore reach significant concentrations in
safely receive b-lactam–based
remains the preferred agent, but amniotic fluid until multiple doses are
regimens for GBS IAP.7
ampicillin is acceptable. administered.73,74 The clinical
effectiveness of clindamycin as IAP
administered a median of 6 hours
IAP and GBS EOD
Alternative Antibiotics for GBS IAP
before delivery was only 22% The pharmacokinetics and
Cefazolin is recommended for GBS
compared to no IAP in an ABCs study pharmacodynamics of ampicillin,
IAP for women with a penicillin
that included women treated with penicillin, and cefazolin suggest that
allergy who are at low risk for
clindamycin during the time periods of effective GBS EOD prophylaxis may be
anaphylaxis7 and has similar
pharmacokinetics and mechanisms of 1998–1999 and 2003–2004.76 achieved within 2 to 4 hours of
maternal administration. The clinical
action as ampicillin. Cefazolin rapidly
Vancomycin is recommended for use effectiveness of different durations of
crosses the placenta and is detected
as GBS IAP for women with GBS IAP was evaluated by using
in cord blood and amniotic fluid at
a penicillin allergy who are at high a data set including 7691 births
levels above the GBS MIC within
risk for anaphylaxis if colonized with collected as part of the ABCs system
20 minutes after maternal
clindamycin-resistant GBS isolates. from 2003 to 2004. In this study, the
administration.67–70
Although authors of older studies administration of different durations
Clindamycin is recommended for GBS using ex vivo perfusion models with of penicillin or ampicillin ($4; 2–,4;
IAP for women with a penicillin placental lobules suggested that or ,2 hours before delivery) were
allergy who are at high risk for vancomycin crosses the placental compared to no administration to
anaphylaxis and who are colonized poorly, authors of a subsequent study evaluate the effectiveness of each in
with GBS known to be susceptible to of 13 women undergoing elective preventing GBS EOD. Although all
clindamycin.7 Group B streptococci cesarean delivery found cord blood regimens were effective compared to
are increasingly resistant to concentrations of vancomycin greater no IAP, administration of IAP at $4
clindamycin as well as to macrolide than the GBS MIC within 30 minutes hours before delivery was most
antibiotics such as erythromycin. In of maternal drug administration.77,78 effective in preventing GBS EOD.76
reports from the ABCs from 2016, Authors of recent studies compared This study was limited by the fact that
authors found that 42% of GBS cord blood vancomycin 85% of the cases occurred among
isolates were resistant to clindamycin concentrations after vancomycin was infants born to women who screened
and 54% were resistant to administered to pregnant women negative for GBS or whose GBS status
erythromycin.71 Because of both poor using standard dosing and maternal was unknown, meaning that in most
placental kinetics and high levels of weight-based dosing.79,80 Therapeutic cases, GBS IAP was administered in

PEDIATRICS Volume 144, number 2, August 2019 5


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
response to risk factors and not as colonized with group B streptococci at of neonates with suspected or proven
prophylaxis for known maternal GBS 1 week and/or 1 month of age.82 The early-onset bacterial sepsis.83,84 In
colonization. Despite this limitation, authors also observed that 6.5% of summary, at this time, evidence
this study, combined with the time- infants born to mothers who screened supports the following:
dependent bactericidal mechanism of negative for GBS were colonized at • The ACOG provides guidance
action of b-lactam antibiotics, 1 week and/or 1 month of age, likely regarding intraamniotic infection85;
supports the effectiveness of as a result of horizontal transmission neonatal risk assessment can be
ampicillin and penicillin administered from newly colonized mothers as well informed by this guidance. The
at least 4 hours before delivery. No as from other contacts. definitive diagnosis of intraamniotic
data specifically inform the clinical infection is that made by amniotic
effectiveness of cefazolin IAP, but the RISK ASSESSMENT FOR EARLY-ONSET fluid Gram-stain and/or culture or
pharmacokinetics and mechanism of GBS INFECTION by placental histopathologic testing.
bacterial killing action for cefazolin are Such clear diagnostic information
similar enough to those of penicillin Because the pathogenesis of GBS EOD
begins with vertical transmission of will rarely be available at the time of
and ampicillin that the administration delivery for infants born at or near
of cefazolin can be considered to group B streptococci from mother to
fetus and newborn infant, the term. Suspected intraamniotic
provide adequate prophylaxis against infection is defined as a single
GBS EOD. Although data regarding the strongest predictor of GBS EOD is
maternal GBS colonization. Other maternal intrapartum temperature
pharmacokinetics of clindamycin and $39.0°C or maternal temperature of
vancomycin have been published, factors that are important to the
pathogenesis of GBS EOD, such as the 38.0°C to 38.9°C in combination with
evidence regarding their clinical 1 or more of maternal leukocytosis,
efficacy is more limited. Therefore, for virulence of the maternal colonizing
isolate and the presence of maternal purulent cervical drainage, or fetal
the purpose of newborn GBS EOD risk
serotype-specific protective antibody, tachycardia. Recognizing the
assessment, when non–b-lactam
cannot be known to the physician at uncertainties surrounding the
antibiotics of any duration are
the time of neonatal risk assessment. diagnosis of intraamniotic infection,
administered for GBS IAP, such
The remaining clinical risk factors for the ACOG recommends that
treatment should be considered as not
GBS EOD are the same as those for intrapartum antibiotic therapy be
fully adequate in neonatal risk
EOS caused by other common administered whenever intra-
calculation.
bacterial causes of EOS, including amniotic infection is diagnosed or
IAP and GBS LOD gestational age at birth, intraamniotic suspected and should be considered
infection, and duration of ROM. The when otherwise unexplained
There is no epidemiological evidence
administration of GBS IAP and the isolated maternal temperature 38.0°
to suggest a protective effect of GBS
administration of intrapartum C to 38.9°C is present.85
IAP for the prevention of GBS LOD.
This observation is likely attributable antibiotics in response to obstetric • The routine measurement of
to the dynamic nature of GBS concern for intraamniotic infection complete blood cell counts or
colonization. Early studies of IAP both decrease this risk. The newborn inflammatory markers such as
demonstrated initial maternal vaginal infant’s condition at birth and C-reactive protein alone in
and/or rectal GBS clearance with IAP, evolving condition over the first 12 to newborn infants to determine risk
followed by recolonization with group 24 hours after birth are strong of GBS EOD is not justified given
B streptococci within 24 to 48 hours predictors of early-onset infection the poor test performance of these
after birth.30 Authors of a study in attributable to any pathogen.12,13 in predicting what is currently
Italy found that approximately 25% of Previous guidance on GBS EOD risk a low-incidence disease.86–89
infants born to mothers with GBS assessment presented challenges to • Newborn clinical illness consisting
colonization who received GBS IAP physicians because of practical of abnormal vital signs (eg,
were colonized by 1 month of age. difficulties in establishing the tachycardia, tachypnea, and/or
Molecular analysis revealed the infant obstetric diagnosis of maternal temperature instability),
colonization was attributable to the chorioamnionitis or intraamniotic supplemental oxygen requirement
same strain colonizing the mother infection, absence of guidance on what and/or need for continuous
antepartum.81 Similarly, authors of defines abnormal laboratory test positive airway pressure,
a longitudinal cohort study conducted results in the newborn infant, and mechanical ventilation, or blood
in Japan from 2014 to 2015 observed a lack of clear definitions for newborn pressure support can be used to
that among neonates born to mothers clinical illness. Each of these concerns predict early-onset infection.13
with GBS colonization who received are addressed in detail in the current There is no evidence that
IAP, approximately 20% were AAP clinical reports on management hypoglycemia occurring in isolation

6 FROM THE AMERICAN ACADEMY OF PEDIATRICS


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
in otherwise well-appearing infants of mothers who screened negative for current era of widespread use of GBS
is a risk factor for GBS EOD or EOS. GBS because of changes in maternal IAP, the clinical approach to risk of
A newborn’s clinical condition often colonization status during the GBS EOD should be the same as that
evolves in the hours after birth, and interval from screening to for all bacterial causes of early-onset
physicians must exercise judgment presentation for delivery or because infection. As addressed in the AAP
to distinguish transitional instability of an incorrect technique in obtaining clinical reports on management of
from signs of clinical illness. vaginal and rectal screening cultures neonates with suspected or proven
Clinicians should recognize that GBS or in laboratory processing. Studies early-onset bacterial sepsis,83,84 risk of
EOD can occur among term infants comparing antepartum culture to early-onset infection should be
born to mothers who have screened intrapartum culture or intrapartum considered separately for infants born
negative for GBS. Authors of 1 single- molecular identification techniques at or near term ($35 weeks’
center study with policies mandating reveal that approximately 7% to 8% gestation) and those preterm (#34 6/
universal screening-based GBS IAP of women who screen negative for 7 weeks’ gestation). A summary of the
found that over an 8-year period, 17 GBS will be identified as positive for management strategies provided in
GBS EOD cases occurred among term GBS at the time of delivery.27,91 In the previous AAP reports are
infants and 14 of 17 (82.4%) of the some proportion of these cases, provided here.
mothers had screened negative for additional risk factors for EOS were
GBS.90 Multistate ABCs data in present, but GBS EOD can develop in There Are 3 Current Approaches to
2003–2004 identified 189 cases of newborn infants without additional Risk Assessment Among Infants Born
GBS EOD among term infants and risk, and these will only be identified at ‡35 Weeks’ Gestation
determined that 116 of 189 (61.4%) when they develop signs of illness. • Categorical risk assessment:
occurred among infants born to Nonetheless, most infants who Categorical risk factor assessment
women who screened negative for develop GBS EOD will do so in the uses risk factor threshold values to
GBS.37 GBS EOD may occur in infants setting of specific risk factors. In the identify infants at increased risk for

FIGURE 1
Options for EOS risk assessment among infants born $35 weeks’ gestation. A, Categorical risk assessment. B, Neonatal Early-Onset Sepsis Calculator. The
screenshot of the Neonatal Early-Onset Sepsis Calculator (https://neonatalsepsiscalculator.kaiserpermanente.org/) was used with permission from
Kaiser-Permanente Division of Research. C, Enhanced observation. a Consider lumbar puncture and CSF culture before initiation of empiric antibiotics for
infants who are at the highest risk of infection, especially those with critical illness. Lumbar puncture should not be performed if the infant’s clinical
condition would be compromised, and antibiotics should be administered promptly and not deferred because of procedure delays. b Adequate GBS IAP is
defined as the administration of penicillin G, ampicillin, or cefazolin $4 hours before delivery.

PEDIATRICS Volume 144, number 2, August 2019 7


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
GBS EOD (Fig 1A). Different versions recommended clinical actions to be identify infants who may be at
of this approach have been published taken at specific levels of predicted increased risk of infection. Among
since 1996 and are based on risk. The models begin with the term infants, good clinical
infection epidemiology and expert previous probability of infection, condition at birth is associated with
opinion. For the purpose of and because they predict all an approximately 60% to 70%
categorical risk assessment, maternal bacterial causes of EOS and not only reduction in risk for early-onset
intrapartum temperature $38.0°C is GBS EOD, physicians in the United infection.13 Under this approach,
used as a surrogate for intraamniotic States should enter a previous infants who appear ill at birth and
infection. The administration of probability of 0.5/1000, unless local those who develop signs of illness
penicillin G, ampicillin, or cefazolin incidence of EOS is known to differ over the first 48 hours after birth
$4 hours before delivery is from the national incidence of EOS are treated empirically with
considered adequate GBS IAP; other among term infants. The models do antibiotics.93–95 One center
antibiotics or other durations of account for the content and timing reported the use of clinical
treatment ,4 hours are considered of GBS-specific IAP. When using observation for initially well-
inadequate when using this these models, only penicillin, appearing infants born at $34 to
approach. Substantial data have been ampicillin, or cefazolin should be 35 weeks’ gestation to mothers
reported on the impact of using considered as “GBS-specific with the obstetric diagnosis of
categorical risk factors to manage the antibiotics.” The administration of chorioamnionitis. Whether
risk of GBS EOD.3,14,36,40 However, clindamycin or vancomycin alone observed in a setting with
the risk is highly variable among the for IAP for any duration is currently continuous monitoring or with
newborn infants recommended to recommended to be entered as “no serial examinations during
receive empirical treatment in this antibiotics.” Because the models maternal-infant couplet care, this
approach, ranging from slightly were developed to predict risk of all center ultimately administered
bacterial causes of EOS (and not just empirical antibiotics to 5% to 12%
lower than the baseline population
GBS EOD), and because these of such infants.94,95 Centers that
risk to significantly higher,
models account for other antibiotic adopt this approach will need to
depending on the gestational age,
types and indications for establish processes to ensure serial,
duration of ROM, and timing and
structured, documented physical
content of administered intrapartum intrapartum antibiotic
assessments and develop clear
antibiotics. Consequently, a limitation administration, “GBS specific
criteria for additional evaluation
of this approach is that categorical antibiotics .2 hours prior to birth”
and empirical antibiotic
management will result in empirical is 1 of the calculator variables. The
administration. Physicians and
treatment of many relatively low-risk 2-hour timing is used because
families must understand that the
newborn infants. multiple factors in addition to GBS
identification of initially well-
• Multivariate risk assessment (the IAP are considered when using the
appearing infants who develop
Neonatal Early-Onset Sepsis multivariate models in the Neonatal
clinical illness is not a failure of
Calculator): Multivariate risk Early-Onset Sepsis Calculator. Used
care, but rather an anticipated
assessment integrates the individual in this manner, threshold risk
outcome of this approach to GBS
infant’s combination of risk factors estimates prompting enhanced
EOD risk management.
and the newborn infant’s clinical clinical observation or blood culture
condition to estimate an individual and empirical antibiotic therapy For Infants Born at £34 Weeks’
infant’s risk of EOS, including GBS have been prospectively validated in Gestation, the Optimal Approach to
large newborn cohorts.14,92 Centers Risk Assessment for All EOS Should
EOD. Predictive models based on
that opt for this approach to risk Be Applied to Risk of GBS EOD
gestational age at birth, highest
maternal intrapartum temperature, assessment should develop methods The circumstances of preterm birth
maternal GBS colonization status, to calculate the risk estimate for all may provide the best current
duration of ROM, and type and newborn infants born at $35 approach to GBS EOD management for
duration of intrapartum antibiotic weeks’ gestation and will need to preterm infants (Fig 2). Clinicians may
therapies have been developed and develop a structured approach to adopt one of the following strategies
validated.13,38 These models are close clinical observation of infants to develop institutional approaches
available as a Web-based Neonatal at specific levels of estimated risk. best suited to their local resources and
Early-Onset Sepsis Calculator • Risk assessment based on newborn structure of care (Fig 1).
(Fig 1B) (https:// clinical condition: A final approach • Preterm infants at highest risk for
neonatalsepsiscalculator. to GBS EOD risk assessment is to EOS: Infants born preterm because
kaiserpermanente.org) that includes rely on clinical signs of illness to of cervical insufficiency, preterm

8 FROM THE AMERICAN ACADEMY OF PEDIATRICS


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
labor, PROM, intraamniotic infection, growth restriction), (2) birth by any other concern for infection
and/or acute and otherwise cesarean delivery, and (3) absence arises during the process of
unexplained onset of nonreassuring of labor, attempts to induce labor, delivery, the infant should be
fetal status are at the highest risk of or any ROM before delivery. managed as recommended above
EOS and GBS EOD. The Acceptable initial approaches to for preterm infants at higher risk
administration of GBS IAP may these infants include (1) no for GBS EOD. Otherwise, an
decrease the risk of infection among laboratory evaluation and no acceptable approach to these
these infants, but the most empirical antibiotic therapy or (2) infants is close observation for
reasonable approach to these blood culture and clinical those infants who are well
infants is to obtain a blood culture monitoring. For infants who do not appearing at birth and to obtain
and start empirical antibiotic improve after initial stabilization a blood culture and to initiate
treatment. A lumbar puncture for and/or those who have severe antibiotic therapy for infants with
culture and analysis of CSF should systemic instability, the respiratory and/or cardiovascular
be considered in clinically ill infants administration of empirical instability after birth.
when there is a high suspicion for antibiotics may be reasonable but
GBS EOD, unless the procedure will is not mandatory. CLINICAL PRESENTATION AND
compromise the neonate’s clinical • Infants delivered for maternal and/ TREATMENT OF GBS INFECTION
condition. or fetal indications but who are Newborn infants with GBS EOD may
• Preterm infants at lower risk for ultimately born by vaginal or present with signs of illness ranging
EOS: Preterm infants at lowest risk cesarean delivery after efforts to from tachycardia, tachypnea, or
for all EOS and for GBS EOD are induce labor and/or with ROM lethargy to severe cardiorespiratory
those born under circumstances before delivery are subject to failure, persistent pulmonary
that include all of these criteria96, factors associated with the hypertension of the newborn, and
97
: (1) maternal and/or fetal pathogenesis of GBS EOD. If the perinatal encephalopathy. GBS LOD
indications for preterm birth (such mother has an indication for GBS most commonly occurs as bacteremia
as maternal preeclampsia or other IAP and adequate IAP (penicillin, without a focus and is often
noninfectious medical illness, ampicillin, or cefazolin $4 hours characterized by fever ($38°C) as
placental insufficiency, or fetal before delivery) is not given or if well as lethargy, poor feeding,

FIGURE 2
EOS risk assessment among infants born #34 weeks’ gestation. a Intraamniotic infection should be considered when a pregnant woman presents with
unexplained decreased fetal movement and/or there is sudden and unexplained poor fetal testing. b Lumbar puncture and CSF culture should be
performed before initiation of empiric antibiotics for infants who are at the highest risk of infection unless the procedure would compromise the infant’s
clinical condition. Antibiotics should be administered promptly and not deferred because of procedural delays. c Adequate GBS IAP is defined as the
administration of penicillin G, ampicillin, or cefazolin $4 hours before delivery. d For infants who do not improve after initial stabilization and/or those
who have severe systemic instability, the administration of empiric antibiotics may be reasonable but is not mandatory.

PEDIATRICS Volume 144, number 2, August 2019 9


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
irritability, tachypnea, grunting, or Empirical and Definitive Treatment continuously hospitalized preterm
apnea. Infants with LOD meningitis Antibiotics initiated because of infants beyond 72 hours of age is
can also have nonspecific signs such concern for GBS EOD are the same as guided by multiple factors, including
as irritability, poor feeding, vomiting, those for all bacterial causes of EOS the presence of central venous
and temperature instability as well as until the results of cultures are catheters and local hospital
signs suggestive of central nervous available. GBS neonatal disease microbiology. The empirical choice
systems involvement such as bulging isolates remain susceptible to for such infants should include an
fontanelle or seizures. Focal b-lactam antibiotics, although ABCs antibiotic to which group B
syndromes such as pneumonia, bone has identified rare isolates of group B streptococci are susceptible, such as
or joint infection, cellulitis, or adenitis streptococci with mutations in a b-lactam, cephalosporin, or
often have findings that point to the penicillin-binding proteins leading to vancomycin. When group B
site of infection. Osteomyelitis elevated MICs (nonsusceptibility) to streptococci are identified in culture,
frequently is insidious and may not b-lactam antibiotics.99 Ampicillin, penicillin G is the drug of choice, with
be associated with fever. together with an aminoglycoside, is ampicillin as an acceptable
Complications are common for the primary recommended therapy alternative therapy.
meningitis and can include for infants up to 7 days of age. The
neurodevelopment impairment, empirical addition of broader- See Table 1 for dose and interval
hearing loss, persistent seizure spectrum therapy should be guidance. The length of antibiotic
disorders, or cerebrovascular considered if there is strong clinical treatment is generally 10 days for
disease.98 concern for ampicillin-resistant bacteremia without focus and 14 days
infection in a critically ill newborn, for uncomplicated meningitis;
Evaluation for GBS disease is the particularly among neonates with antibiotics should be given
same as that for all forms of sepsis in very low birth weight.100–102 Among intravenously for the entire course.
the newborn and young infant.83,84 previously healthy infants in the Longer therapy is used when there is
Blood culture, lumbar puncture for community, if the infant is not a prolonged or complicated course.
culture and analysis of CSF, and critically ill and there is no evidence Some experts recommend a second
markers of inflammation are of meningitis, ampicillin and lumbar puncture for CSF culture 24
discussed in detail in the AAP clinical ceftazidime together are to 48 hours after the start of
reports on management of neonates recommended as empirical therapy antibiotics. Additional lumbar
with suspected or proven early-onset for those 8 to 28 days of age; punctures and intracranial imaging
bacterial sepsis. The evaluation for ceftriaxone therapy is recommended are advised if there is not resolution
GBS LOD should also include urine under these circumstances for infants of CSF infection, if neurologic
culture. If bone or joint infection is 29 to 90 days of age. For all abnormalities persist, or if focal
suspected, additional studies can previously healthy infants in the deficits develop. Osteoarticular
include radiographs, MRI, and bone community from 8 to 90 days of age, infection should be treated for 3 to
or joint fluid culture. When vancomycin should be added to 4 weeks and ventriculitis should be
meningitis is diagnosed, cranial recommended empirical therapy if treated for at least 4 weeks.
imaging is often useful to assess for there is evidence of meningitis or Consultation with a pediatric
complications such as ventriculitis or critical illness to expand coverage, infectious disease specialist should be
brain abscess. Cultures should including for b-lactam–resistant considered for meningitis and for
include testing for antibiotic Streptococcus pneumoniae. The choice cases with site-specific infection.
susceptibility. of empirical therapy among Audiology testing and ongoing

TABLE 1 Recommended Intravenous Antibiotic Treatment Regimens for Confirmed Early- and Late-Onset GBS Bacteremia and Meningitis
GA #34 wk GA .34 wk
PNA #7 d PNA .7 d PNA #7 d PNA .7 d
Bacteremia
Ampicillin 50 mg/kg every 12 h 75 mg/kg every 12 h 50 mg/kg every 8 h 50 mg/kg every 8 h
Penicillin G 50 000 U/kg every 12 h 50 000 U/kg every 8 h 50 000 U/kg every 12 h 50 000 U/kg every 8 h
Meningitis
Ampicillin 100 mg/kg every 8 h 75 mg/kg every 6 h 100 mg/kg every 8 h 75 mg/kg q 6 h
Penicillin G 150 000 U/kg every 8 h 125 000 U/kg every 6 h 150 000 U/kg every 8 h 125 000 U/kg every 6 h
Adapted from Table 4.2. Antibacterial Drugs for Neonates (,28 Postnatal Days of Age). In: Kimberlin DW, Brady MT, Jackson MA, Long SS, eds. Red Book: 2018 Report of the Committee on
Infectious Diseases. 31st ed. Itasca, IL: American Academy of Pediatrics; 2018:915-919. GA, gestational age; PNA, postnatal age.

10 FROM THE AMERICAN ACADEMY OF PEDIATRICS


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
audiologic monitoring, if indicated, of the gut microbiota develops and malignancy.15,120 Furthermore, GBS
should be arranged before discharge. diversifies from birth through early disease is a worldwide problem,
childhood, and disruption of the particularly among resource-limited
Multiple Births microbiota during this critical countries where IAP is not a readily
When invasive GBS infection occurs period may have enduring health available preventive strategy.
in an infant who is 1 of multiple consequences. Perinatal antibiotic Effective, multivalent vaccines
births, the birth siblings should be administration is associated with administered to pregnant women and
observed carefully for signs of abnormal host development in at-risk nonpregnant adults could
infection and treated empirically if animal models.106–108 Human potentially prevent many of these
signs of illness occur. There is no epidemiological studies have associated issues. Preclinical and human phase I
evidence to support full antibiotic early infant antibiotic exposures with and II studies have been completed
treatment courses in the absence of increased risks of atopic and allergic revealing the safety and
confirmed GBS disease. disorders as well as increases in early immunogenicity of glycoconjugate
childhood weight gain.109–112 Several GBS vaccines.121–123 Current ABCs
Recurrent GBS Disease human studies report differences in the reveals that 99% of infections are
composition of the maternal vaginal113 caused by 6 GBS serotypes,
Recurrent neonatal and young infant
and infant gut microbiome associated suggesting that a hexavalent vaccine
GBS disease can occur after
with the use of IAP, with effects on both could be widely effective.6
completed appropriate treatment of
the diversity and richness of identified
the primary infection.103,104 Authors
species.114–119 The differences observed
of a population-based study SUMMARY OF RECOMMENDATIONS
vary with the mode of delivery and
conducted in Japan53 during 2011 to
duration of breastfeeding; differences 1. The AAP supports the maternal
2015 found that recurrence occurred
were detected as long as 12 months policies and procedures for the
in 2.8% of cases of neonatal GBS
after birth in 1 study.115 Changes in the prevention of perinatal GBS
infection. Recurrent cases were
initial constitution of neonatal gut disease as recommended by
identified 3 to 54 days after
microbiota in response to IAP are not the ACOG.
completion of the therapy for the first
unexpected; GBS IAP is administered
occurrence. Recurrent cases are 2. For the purpose of neonatal
with the intent of altering the content of
generally caused by the same GBS management, the administration of
the newborn infant’s initial microflora
serotype that caused the primary intrapartum penicillin G, ampicillin,
exposure at delivery to decrease
infection, and persistent mucosal or cefazolin can provide adequate
colonization with a significant neonatal
colonization and poor neonatal IAP against neonatal early-onset GBS
pathogen. Whether the secondary
antibody responses to the first disease. For women at high risk of
effects of IAP on the microbiome
infection likely contribute to the anaphylaxis to b-lactam antibiotics,
influence short- and long-term
pathogenesis of recurrent infection. clindamycin and vancomycin should
childhood health outcomes is unknown
Recurrent disease is not preventable be administered as recommended by
and an area of active investigation.
by extension of recommended the ACOG and will likely provide
antibiotic courses nor by the addition GBS Vaccine Development some protection against GBS EOD in
of rifampin to eradicate mucosal exposed newborn infants. However,
Multiple gaps remain in GBS disease
colonization.105 Parents should be there is currently insufficient clinical
prevention. Neonatal EOD continues
counseled about the possibility of efficacy evidence to consider the
to occur in the United States,
recurrent GBS disease after treatment administration of these antibiotics
primarily among preterm infants,
of both GBS EOD and LOD. equivalent to b-lactam antibiotics for
term infants born to women with
the purpose of neonatal risk
cultures negative for antenatal GBS,
assessment.
FUTURE DIRECTIONS and infants born to mothers with
reported b-lactam allergy that 3. Risk assessment for early-onset
Intrapartum Antibiotics, the precludes maximally effective IAP. GBS disease should follow the
Microbiome, and Childhood Health The incidence of GBS LOD has not general principles established in
GBS IAP and the administration of been affected by IAP. In addition, the AAP clinical reports on
intrapartum antibiotics because of group B Streptococcus is a cause of management of neonates with
concern for maternal intraamniotic stillbirth and of invasive infection suspected or proven early-onset
infection combined result in among pregnant and postpartum bacterial sepsis.83,84 These
approximately 30% of pregnant women as well as among the elderly principles include the following:
women receiving antibiotics around and those whose health is ○ separate consideration of infants
the time of delivery. The composition compromised by diabetes or born at $35 0/7 weeks’

PEDIATRICS Volume 144, number 2, August 2019 11


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
gestation and those born at #34 Ivan Hand, MD, FAAP Karen M. Farizo, MD – US Food and Drug
6/7 weeks’ gestation; Ira Adams-Chapman, MD, FAAP Administration
Brenda Poindexter, MD, FAAP Marc Fischer, MD, FAAP – Centers for Disease
○ infants born at $35 0/7 weeks’ Dan L. Stewart, MD, FAAP Control and Prevention
gestation may be assessed for risk Susan W. Aucott, MD, FAAP Natasha B. Halasa, MD, MPH, FAAP –
of early-onset GBS infection with Karen M. Puopolo, MD, PhD, FAAP Pediatric Infectious Diseases Society
a categorical algorithm by using Jay P. Goldsmith, MD, FAAP Nicole Le Saux, MD, FRCP(C) – Canadian
Meredith Mowitz, MD, FAAP Paediatric Society
multivariate models such as the Kristi Watterberg, MD, FAAP, Immediate Past Scot B. Moore, MD, FAAP – Committee on
Neonatal Early-Onset Sepsis Chairperson Practice Ambulatory Medicine
Calculator or with enhanced Neil S. Silverman, MD – American College of
clinical observation; and Obstetricians and Gynecologists
LIAISONS Jeffrey R. Starke, MD – FAAP, American
○ infants born at #34 6/7 weeks’ Thoracic Society
Timothy Jancelewicz, MD – American
gestation are at highest risk for Academy of Pediatrics Section on Surgery James J. Stevermer, MD, MSPH, FAAFP –
early-onset infection from all Michael Narvey, MD – Canadian Pediatric American Academy of Family Physicians
causes, including group B Society Kay M. Tomashek, MD, MPH, DTM – National
Russell Miller, MD – American College of Institutes of Health
streptococci, and may be best
approached by using the Obstetricians and Gynecologists
RADM Wanda Barfield, MD, MPH – Centers
circumstances of preterm birth STAFF
for Disease Control and Prevention
to determine management. Erin Keels, DNP, APRN, NNP-BC – National Jennifer M. Frantz, MPH
4. Early-onset GBS infection is Association of Neonatal Nurses
diagnosed by blood or CSF culture. ACKNOWLEDGMENTS
Common laboratory tests such as STAFF The American Academy of Pediatrics thanks
the complete blood cell count and Stephanie Schrag, DPhil, Epidemiology Team
Jim Couto, MA
C-reactive protein do not perform Lead, Respiratory Diseases Branch, Centers
well in predicting early-onset for Disease Control and Prevention, and
infection, particularly among well- COMMITTEE ON INFECTIOUS DISEASES, Carol Baker, MD, McGovern Medical School,
2018–2019 University of Texas-Houston, for their
appearing infants at lowest partnership and contributions to the
baseline risk of infection. Yvonne A. Maldonado, MD, FAAP, development of this clinical report.
Chairperson
5. Evaluation for late-onset GBS
Theoklis E. Zaoutis, MD, MSCE, FAAP, Vice
disease should be based on clinical Chairperson
signs of illness in the infant. Ritu Banerjee, MD, PhD, FAAP ABBREVIATIONS
Diagnosis is based on the isolation Elizabeth D. Barnett MD, FAAP AAP: American Academy of
of group B streptococci from blood, James D. Campbell, MD, MS, FAAP
Pediatrics
Jeffrey S. Gerber, MD, PhD, FAAP
CSF, or other normally sterile sites. ABCs: Active Bacterial Core
Athena P. Kourtis, MD, PhD, MPH, FAAP
Late-onset GBS disease occurs Ruth Lynfield, MD, FAAP surveillance
among infants born to mothers who Flor M. Munoz, MD, MSc, FAAP ACOG: American College of
had positive GBS screen results as Dawn Nolt, MD, MPH, FAAP Obstetricians and
well as those who had negative Ann-Christine Nyquist, MD, MSPH, FAAP
Gynecologists
Sean T. O’Leary, MD, MPH, FAAP
screen results during pregnancy. CDC: Centers for Disease Control
Mark H. Sawyer, MD, FAAP
Adequate IAP does not protect William J. Steinbach, MD, FAAP and Prevention
infants from late-onset GBS disease. Ken Zangwill, MD, FAAP CSF: cerebrospinal fluid
6. Empirical antibiotic therapy for EOD: early-onset disease
early-onset and late-onset GBS EX OFFICIO EOS: early-onset sepsis
disease differs by postnatal age at GBS: group B streptococcal
David W. Kimberlin, MD, FAAP – Red Book
the time of evaluation. Penicillin G Editor
IAP: intrapartum antibiotic
is the preferred antibiotic for Henry H. Bernstein, DO, MHCM, FAAP – Red prophylaxis
definitive treatment of GBS disease Book Online Associate Editor LOD: late-onset disease
in infants; ampicillin is an H. Cody Meissner, MD, FAAP – Visual Red MIC: minimum inhibitory
Book Associate Editor concentration
acceptable alternative.
NAAT: nucleic acid
LIAISONS amplification test
COMMITTEE ON FETUS AND NEWBORN, PROM: prelabor rupture of
Amanda C. Cohn, MD, FAAP – Centers for
2018–2019 Disease Control and Prevention membranes
James Cummings, MD, MS, FAAP, Jamie Deseda-Tous, MD – Sociedad ROM: rupture of membranes
Chairperson Latinoamericana de Infectología Pediátrica

12 FROM THE AMERICAN ACADEMY OF PEDIATRICS


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
FINANCIAL DISCLOSURE: The authors have indicated they have no financial relationships relevant to this article to disclose.
FUNDING: No external funding.
POTENTIAL CONFLICT OF INTEREST: The authors have indicated they have no potential conflicts of interest to disclose.

REFERENCES
1. Centers for Disease Control and Kimberlin DW, Brady MT, Jackson MA, 6 years in France. Acta Paediatr. 2010;
Prevention. Prevention of perinatal Long SS, eds. Red Book: 2018 99(1):47–51
group B streptococcal disease: Report of the Committee on
18. Boyer KM, Gadzala CA, Burd LI, Fisher
a public health perspective [published Infectious Diseases. 31st ed. Itasca,
DE, Paton JB, Gotoff SP. Selective
correction appears in MMWR Morb IL: American Academy of Pediatrics;
intrapartum chemoprophylaxis of
Mortal Wkly Rep. 1996;45(31):679]. 2018:762–768
neonatal group B streptococcal early-
MMWR Recomm Rep. 1996;45(RR-7):
9. Schrag SJ, Farley MM, Petit S, et al. onset disease. I. Epidemiologic
1–24
Epidemiology of invasive early-onset rationale. J Infect Dis. 1983;148(5):
2. Schrag S, Gorwitz R, Fultz-Butts K, neonatal sepsis, 2005 to 2014. 795–801
Schuchat A. Prevention of perinatal Pediatrics. 2016;138(6):e20162013
19. Baker CJ, Barrett FF. Transmission of
group B streptococcal disease.
10. Weston EJ, Pondo T, Lewis MM, et al. The group B streptococci among parturient
Revised guidelines from CDC. MMWR
burden of invasive early-onset neonatal women and their neonates. J Pediatr.
Recomm Rep. 2002;51(RR-11):1–22
sepsis in the United States, 2005-2008. 1973;83(6):919–925
3. Schrag SJ, Zell ER, Lynfield R, et al; Pediatr Infect Dis J. 2011;30(11):
Active Bacterial Core Surveillance 937–941 20. Benitz WE, Gould JB, Druzin ML. Risk
Team. A population-based comparison factors for early-onset group B
11. Baker CJ. Early onset group B streptococcal sepsis: estimation of
of strategies to prevent early-onset
streptococcal disease. J Pediatr. 1978; odds ratios by critical literature review.
group B streptococcal disease in
93(1):124–125 Pediatrics. 1999;103(6). Available at:
neonates. N Engl J Med. 2002;347(4):
233–239 12. Escobar GJ, Li DK, Armstrong MA, et al. www.pediatrics.org/cgi/content/full/
Neonatal sepsis workups in infants 103/6/e77
4. Verani JR, McGee L, Schrag SJ; Division
./=2000 grams at birth: a population- 21. Goldenberg RL, Hauth JC, Andrews WW.
of Bacterial Diseases, National Center
based study. Pediatrics. 2000;106(2 pt Intrauterine infection and preterm
for Immunization and Respiratory
1):256–263 delivery. N Engl J Med. 2000;342(20):
Diseases, Centers for Disease Control
and Prevention (CDC). Prevention of 13. Escobar GJ, Puopolo KM, Wi S, et al. 1500–1507
perinatal group B streptococcal Stratification of risk of early-onset 22. Gibbs RS, Roberts DJ. Case records of
disease--revised guidelines from CDC, sepsis in newborns $ 34 weeks’ the Massachusetts General Hospital.
2010. MMWR Recomm Rep. 2010;59(RR- gestation. Pediatrics. 2014;133(1):30–36 Case 27-2007. A 30-year-old pregnant
10):1–36 14. Kuzniewicz MW, Puopolo KM, Fischer A, woman with intrauterine fetal
5. Mukhopadhyay S, Taylor JA, Von et al. A quantitative, risk-based death. N Engl J Med. 2007;357(9):
Kohorn I, et al. Variation in sepsis approach to the management of 918–925
evaluation across a national network neonatal early-onset sepsis. JAMA 23. Nan C, Dangor Z, Cutland CL, Edwards
of nurseries. Pediatrics. 2017;139(3): Pediatr. 2017;171(4):365–371 MS, Madhi SA, Cunnington MC. Maternal
e0162845 group B Streptococcus-related
15. Seale AC, Bianchi-Jassir F, Russell NJ,
6. Nanduri SA, Petit S, Smelser C, et al. et al. Estimates of the burden of group stillbirth: a systematic review. BJOG.
Epidemiology of invasive early-onset B streptococcal disease worldwide for 2015;122(11):1437–1445
and late-onset group B streptococcal pregnant women, stillbirths, and 24. Russell NJ, Seale AC, O’Driscoll M, et al;
disease in the United States, 2006 to children. Clin Infect Dis. 2017;65(suppl_ GBS Maternal Colonization Investigator
2015: multistate laboratory and 2):S200–S219 Group. Maternal colonization with
population-based surveillance. JAMA group B Streptococcus and serotype
16. Hussain SM, Luedtke GS, Baker CJ,
Pediatr. 2019;173(3):224–233 distribution worldwide: systematic
Schlievert PM, Leggiadro RJ. Invasive
7. American College of Obstetricians and group B streptococcal disease in review and meta-analyses. Clin Infect
Gynecologists. Prevention of group B children beyond early infancy. Pediatr Dis. 2017;65(suppl_2):S100–S111
streptococcal early-onset disease in Infect Dis J. 1995;14(4):278–281
25. Campbell JR, Hillier SL, Krohn MA,
newborns: ACOG Committee Opinion,
17. Guilbert J, Levy C, Cohen R, Delacourt C, Ferrieri P, Zaleznik DF, Baker CJ. Group
Number 782. Obstet Gynecol. 2019;
Renolleau S, Flamant C; Bacterial B streptococcal colonization and
134(1):e19–e40
meningitis group. Late and ultra late serotype-specific immunity in pregnant
8. American Academy of Pediatrics. onset Streptococcus B meningitis: women at delivery. Obstet Gynecol.
Group B streptococcal infections. In: clinical and bacteriological data over 2000;96(4):498–503

PEDIATRICS Volume 144, number 2, August 2019 13


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
26. Buchan BW, Faron ML, Fuller D, Davis TE, Study Group. Multistate case-control streptococcal disease. J Infect Dis.
Mayne D, Ledeboer NA. Multicenter study of maternal risk factors for 2014;209(5):781–788
clinical evaluation of the Xpert GBS LB neonatal group B streptococcal
46. Collins A, Weitkamp JH, Wynn JL. Why
assay for detection of group B disease. Pediatr Infect Dis J. 1994;13(7):
are preterm newborns at increased
Streptococcus in prenatal screening 623–629
risk of infection? Arch Dis Child Fetal
specimens. J Clin Microbiol. 2015;53(2):
36. Schuchat A, Zywicki SS, Dinsmoor MJ, Neonatal Ed. 2018;103(4):F391–F394
443–448
et al. Risk factors and opportunities for
27. Young BC, Dodge LE, Gupta M, Rhee JS, prevention of early-onset neonatal 47. Adair CE, Kowalsky L, Quon H, et al. Risk
Hacker MR. Evaluation of a rapid, real- sepsis: a multicenter case-control factors for early-onset group B
time intrapartum group B study. Pediatrics. 2000;105(1 pt 1):21–26 streptococcal disease in neonates:
streptococcus assay. Am J Obstet a population-based case-control study.
37. Van Dyke MK, Phares CR, Lynfield R, CMAJ. 2003;169(3):198–203
Gynecol. 2011;205(4):372.e1–372.e6
et al. Evaluation of universal antenatal
28. Hansen SM, Uldbjerg N, Kilian M, screening for group B streptococcus. 48. Pintye J, Saltzman B, Wolf E, Crowell CS.
Sørensen UB. Dynamics of N Engl J Med. 2009;360(25):2626–2636 Risk factors for late-onset group B
Streptococcus agalactiae colonization streptococcal disease before and after
in women during and after pregnancy 38. Puopolo KM, Draper D, Wi S, et al. implementation of universal screening
and in their infants. J Clin Microbiol. Estimating the probability of neonatal and intrapartum antibiotic prophylaxis.
2004;42(1):83–89 early-onset infection on the basis of J Pediatric Infect Dis Soc. 2016;5(4):
maternal risk factors. Pediatrics. 2011; 431–438
29. Meyn LA, Moore DM, Hillier SL, Krohn 128(5). Available at: www.pediatrics.
MA. Association of sexual activity with org/cgi/content/full/128/5/e1155 49. Berardi A, Rossi C, Lugli L, et al; GBS
colonization and vaginal acquisition of Prevention Working Group, Emilia-
group B Streptococcus in nonpregnant 39. Zaleznik DF, Rench MA, Hillier S, et al. Romagna. Group B streptococcus late-
women. Am J Epidemiol. 2002;155(10): Invasive disease due to group B onset disease: 2003-2010. Pediatrics.
949–957 Streptococcus in pregnant women and 2013;131(2). Available at: www.
neonates from diverse population pediatrics.org/cgi/content/full/131/2/
30. Yow MD, Mason EO, Leeds LJ, Thompson groups. Clin Infect Dis. 2000;30(2): e361
PK, Clark DJ, Gardner SE. Ampicillin 276–281
prevents intrapartum transmission of 50. Lin FY, Weisman LE, Troendle J, Adams K.
group B streptococcus. JAMA. 1979; 40. Mukhopadhyay S, Dukhovny D, Mao W, Prematurity is the major risk factor for
241(12):1245–1247 Eichenwald EC, Puopolo KM. 2010 late-onset group B streptococcus
perinatal GBS prevention guideline and disease. J Infect Dis. 2003;188(2):
31. Boyer KM, Gadzala CA, Kelly PD, Gotoff SP. resource utilization. Pediatrics. 2014; 267–271
Selective intrapartum chemoprophylaxis 133(2):196–203
of neonatal group B streptococcal 51. Jordan HT, Farley MM, Craig A, et al;
early-onset disease. III. Interruption of 41. Heath PT, Balfour GF, Tighe H, Verlander Active Bacterial Core Surveillance
mother-to-infant transmission. J Infect NQ, Lamagni TL, Efstratiou A; HPA GBS (ABCs)/Emerging Infections Program
Dis. 1983;148(5):810–816 Working Group. Group B streptococcal Network, CDC. Revisiting the need for
disease in infants: a case control study. vaccine prevention of late-onset
32. de Cueto M, Sanchez MJ, Sampedro A, Arch Dis Child. 2009;94(9):674–680
Miranda JA, Herruzo AJ, Rosa-Fraile M. neonatal group B streptococcal
Timing of intrapartum ampicillin and 42. Carstensen H, Christensen KK, Grennert disease: a multistate, population-based
prevention of vertical transmission of L, Persson K, Polberger S. Early-onset analysis. Pediatr Infect Dis J. 2008;
group B streptococcus. Obstet Gynecol. neonatal group B streptococcal 27(12):1057–1064
1998;91(1):112–114 septicaemia in siblings. J Infect. 1988; 52. Bartlett AW, Smith B, George CR, et al.
17(3):201–204 Epidemiology of late and very late onset
33. Russell NJ, Seale AC, O’Sullivan C, et al.
Risk of early-onset neonatal group B 43. Persson K, Bjerre B, Elfström L, group B streptococcal disease: fifteen-
streptococcal disease with maternal Polberger S, Forsgren A. Group B year experience from two Australian
colonization worldwide: systematic streptococci at delivery: high count in tertiary pediatric facilities. Pediatr
review and meta-analyses. Clin Infect urine increases risk for neonatal Infect Dis J. 2017;36(1):20–24
Dis. 2017;65(suppl_2):S152–S159 colonization. Scand J Infect Dis. 1986; 53. Matsubara K, Hoshina K, Kondo M, et al.
18(6):525–531
34. Schuchat A, Oxtoby M, Cochi S, et al. Group B streptococcal disease in
Population-based risk factors for 44. Baker CJ, Kasper DL. Correlation of infants in the first year of life:
neonatal group B streptococcal maternal antibody deficiency with a nationwide surveillance study in
disease: results of a cohort study in susceptibility to neonatal group B Japan, 2011-2015. Infection. 2017;45(4):
metropolitan Atlanta. J Infect Dis. 1990; streptococcal infection. N Engl J Med. 449–458
162(3):672–677 1976;294(14):753–756
54. Zimmermann P, Gwee A, Curtis N. The
35. Schuchat A, Deaver-Robinson K, 45. Baker CJ, Carey VJ, Rench MA, et al. controversial role of breast milk in GBS
Plikaytis BD, Zangwill KM, Mohle-Boetani Maternal antibody at delivery protects late-onset disease. J Infect. 2017;
J, Wenger JD; The Active Surveillance neonates from early onset group B 74(suppl 1):S34–S40

14 FROM THE AMERICAN ACADEMY OF PEDIATRICS


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
55. Le Doare K, Kampmann B. Breast milk independent patient data. Am 75. Wear CD, Towers CV, Brown MS, Weitz B,
and Group B streptococcal infection: J Perinatol. 2016;33(5):510–517 Porter S, Wolfe L. Transplacental
vector of transmission or vehicle for 65. Barber EL, Zhao G, Buhimschi IA, Illuzzi passage of clindamycin from mother to
protection? Vaccine. 2014;32(26): JL. Duration of intrapartum prophylaxis neonate. J Perinatol. 2016;36(11):
3128–3132 and concentration of penicillin G in fetal 960–961
56. Madrid L, Seale AC, Kohli-Lynch M, et al; serum at delivery. Obstet Gynecol. 2008; 76. Fairlie T, Zell ER, Schrag S. Effectiveness
Infant GBS Disease Investigator Group. 112(2 pt 1):265–270 of intrapartum antibiotic prophylaxis
Infant group B streptococcal disease 66. Berardi A, Pietrangiolillo Z, Bacchi for prevention of early-onset group B
incidence and serotypes worldwide: Reggiani ML, et al. Are postnatal streptococcal disease. Obstet Gynecol.
systematic review and meta-analyses. ampicillin levels actually related to the 2013;121(3):570–577
Clin Infect Dis. 2017;65(suppl_2): duration of intrapartum antibiotic 77. Nanovskaya T, Patrikeeva S, Zhan Y,
S160–S172 prophylaxis prior to delivery? A Fokina V, Hankins GD, Ahmed MS.
57. Patras KA, Nizet V. Group B pharmacokinetic study in 120 neonates. Transplacental transfer of vancomycin
streptococcal maternal colonization Arch Dis Child Fetal Neonatal Ed. 2018; and telavancin. Am J Obstet Gynecol.
and neonatal disease: molecular 103(2):F152–F156 2012;207(4):331.e1–331.e6
mechanisms and preventative 67. Brown CEL, Christmas JT, Bawdon RE.
approaches. Front Pediatr. 2018;6:27 78. Laiprasert J, Klein K, Mueller BA,
Placental transfer of cefazolin and Pearlman MD. Transplacental passage
58. Jiang SM, Cieslewicz MJ, Kasper DL, piperacillin in pregnancies remote from of vancomycin in noninfected term
Wessels MR. Regulation of virulence by term complicated by Rh pregnant women. Obstet Gynecol. 2007;
a two-component system in group B isoimmunization. Am J Obstet Gynecol. 109(5):1105–1110
streptococcus. J Bacteriol. 2005;187(3): 1990;163(3):938–943
1105–1113 79. Towers CV, Weitz B. Transplacental
68. Groff SM, Fallatah W, Yang S, et al. Effect passage of vancomycin. J Matern Fetal
59. Tazi A, Bellais S, Tardieux I, Dramsi S, of maternal obesity on maternal-fetal Neonatal Med. 2018;31(8):1021–1024
Trieu-Cuot P, Poyart C. Group B transfer of preoperative cefazolin at
Streptococcus surface proteins as cesarean section. J Pediatr Pharmacol 80. Onwuchuruba CN, Towers CV, Howard
major determinants for meningeal Ther. 2017;22(3):227–232 BC, Hennessy MD, Wolfe L, Brown MS.
tropism. Curr Opin Microbiol. 2012; Transplacental passage of vancomycin
69. Fiore Mitchell T, Pearlman MD,
15(1):44–49 from mother to neonate. Am J Obstet
Chapman RL, Bhatt-Mehta V, Faix RG.
Gynecol. 2014;210(4):352.e1–352.e4
60. Klinzing DC, Ishmael N, Dunning Hotopp Maternal and transplacental
JC, et al. The two-component response pharmacokinetics of cefazolin. Obstet 81. Berardi A, Rossi C, Creti R, et al. Group B
regulator LiaR regulates cell wall stress Gynecol. 2001;98(6):1075–1079 streptococcal colonization in 160 mother-
responses, pili expression and 70. Allegaert K, van Mieghem T, Verbesselt baby pairs: a prospective cohort study.
virulence in group B Streptococcus. R, et al. Cefazolin pharmacokinetics in J Pediatr. 2013;163(4):1099–104.e1
Microbiology. 2013;159(pt 7):1521–1534 maternal plasma and amniotic fluid 82. Toyofuku M, Morozumi M, Hida M, et al.
61. Mu R, Cutting AS, Del Rosario Y, et al. during pregnancy. Am J Obstet Gynecol. Effects of intrapartum antibiotic
Identification of CiaR regulated genes 2009;200(2):170.e1–170.e7 prophylaxis on neonatal acquisition of
that promote group B streptococcal 71. Centers for Disease Control and group B streptococci. J Pediatr. 2017;
virulence and interaction with brain Prevention. Active bacterial core 190:169–173.e1
endothelial cells. PLoS One. 2016;11(4): surveillance (ABCs): bact facts 83. Puopolo KM, Benitz WE, Zaoutis TE;
e0153891 interactive. Available at: https://wwwn. Committee on Fetus and Newborn;
62. Périchon B, Szili N, du Merle L, et al. cdc.gov/BactFacts/index.html. Accessed Committee on Infectious Diseases.
Regulation of PI-2b pilus expression in February 20, 2019 Management of neonates born at $35
hypervirulent Streptococcus agalactiae 72. Knight KM, Thornburg LL, McNanley AR, 0/7 weeks’ gestation with suspected or
ST-17 BM110. PLoS One. 2017;12(1): Hardy DJ, Vicino D, Glantz JC. The effect proven early-onset bacterial sepsis.
e0169840 of intrapartum clindamycin on vaginal Pediatrics. 2018;142(6):e20182894
63. Boyer KM, Gotoff SP. Prevention of early- group B streptococcus colony counts. 84. Puopolo KM, Benitz WE, Zaoutis TE;
onset neonatal group B streptococcal J Matern Fetal Neonatal Med. 2012; Committee on Fetus and Newborn;
disease with selective intrapartum 25(6):747–749 Committee on Infectious Diseases.
chemoprophylaxis. N Engl J Med. 1986; 73. Weinstein AJ, Gibbs RS, Gallagher M. Management of neonates born at #34
314(26):1665–1669 Placental transfer of clindamycin and 6/7 weeks’ gestation with suspected or
gentamicin in term pregnancy. Am proven early-onset bacterial sepsis.
64. Turrentine MA, Colicchia LC, Hirsch E,
J Obstet Gynecol. 1976;124(7):688–691 Pediatrics. 2018;142(6):e20182896
et al. Efficiency of screening for the
recurrence of antenatal group B 74. Philipson A, Sabath LD, Charles D. 85. Committee on Obstetric Practice.
Streptococcus colonization in Transplacental passage of Committee opinion no. 712: intrapartum
a subsequent pregnancy: a systematic erythromycin and clindamycin. N Engl management of intraamniotic infection.
review and meta-analysis with J Med. 1973;288(23):1219–1221 Obstet Gynecol. 2017;130(2):e95–e101

PEDIATRICS Volume 144, number 2, August 2019 15


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
86. Newman TB, Puopolo KM, Wi S, Draper birth weight infants: opportunities for 106. Cox LM, Yamanishi S, Sohn J, et al.
D, Escobar GJ. Interpreting complete antibiotic stewardship. Pediatr Infect Altering the intestinal microbiota
blood counts soon after birth in Dis J. 2017;36(5):477–481 during a critical developmental window
newborns at risk for sepsis. Pediatrics. has lasting metabolic consequences.
97. Puopolo KM, Mukhopadhyay S, Hansen
2010;126(5):903–909 Cell. 2014;158(4):705–721
NI, et al; NICHD Neonatal Research
87. Hornik CP, Benjamin DK, Becker KC, Network. Identification of extremely 107. Cho I, Yamanishi S, Cox L, et al.
et al. Use of the complete blood cell premature infants at low risk for early- Antibiotics in early life alter the murine
count in early-onset neonatal sepsis. onset sepsis. Pediatrics. 2017;140(5): colonic microbiome and adiposity.
Pediatr Infect Dis J. 2012;31(8):799–802 e20170925 Nature. 2012;488(7413):621–626
88. Newman TB, Draper D, Puopolo KM, Wi 98. Tibussek D, Sinclair A, Yau I, et al. Late- 108. Leclercq S, Mian FM, Stanisz AM, et al.
S, Escobar GJ. Combining immature and onset group B streptococcal meningitis Low-dose penicillin in early life induces
total neutrophil counts to predict early has cerebrovascular complications. long-term changes in murine gut
onset sepsis in term and late preterm J Pediatr. 2015;166(5):1187–1192.e1 microbiota, brain cytokines and
newborns: use of the I/T2. Pediatr Infect behavior. Nat Commun. 2017;8:15062
99. Metcalf BJ, Chochua S, Gertz RE Jr, et al;
Dis J. 2014;33(8):798–802
Active Bacterial Core surveillance team. 109. Kummeling I, Stelma FF, Dagnelie PC,
89. Benitz WE. Adjunct laboratory tests in Short-read whole genome sequencing et al. Early life exposure to antibiotics
the diagnosis of early-onset neonatal for determination of antimicrobial and the subsequent development of
sepsis. Clin Perinatol. 2010;37(2): resistance mechanisms and capsular eczema, wheeze, and allergic
421–438 serotypes of current invasive sensitization in the first 2 years of life:
90. Puopolo KM, Madoff LC, Eichenwald EC. Streptococcus agalactiae recovered in the KOALA Birth Cohort Study.
Early-onset group B streptococcal the USA. Clin Microbiol Infect. 2017; Pediatrics. 2007;119(1). Available at:
disease in the era of maternal 23(8):574.e7–574.e14 www.pediatrics.org/cgi/content/full/
screening. Pediatrics. 2005;115(5): 119/1/e225
100. Schrag SJ, Hadler JL, Arnold KE,
1240–1246 Martell-Cleary P, Reingold A, Schuchat 110. Risnes KR, Belanger K, Murk W, Bracken
91. El Helali N, Nguyen JC, Ly A, A. Risk factors for invasive, early-onset MB. Antibiotic exposure by 6 months
Giovangrandi Y, Trinquart L. Diagnostic Escherichia coli infections in the era of and asthma and allergy at 6 years:
accuracy of a rapid real-time widespread intrapartum antibiotic use. findings in a cohort of 1,401 US
polymerase chain reaction assay for Pediatrics. 2006;118(2):570–576 children. Am J Epidemiol. 2011;173(3):
universal intrapartum group B 310–318
101. Puopolo KM, Eichenwald EC. No change
streptococcus screening. Clin Infect Dis. in the incidence of ampicillin-resistant, 111. Metsälä J, Lundqvist A, Virta LJ, Kaila M,
2009;49(3):417–423 neonatal, early-onset sepsis over Gissler M, Virtanen SM. Mother’s and
92. Dhudasia MB, Mukhopadhyay S, 18 years. Pediatrics. 2010;125(5). offspring’s use of antibiotics and infant
Puopolo KM. Implementation of the Available at: www.pediatrics.org/cgi/ allergy to cow’s milk. Epidemiology.
sepsis risk calculator at an academic content/full/125/5/e1031 2013;24(2):303–309
birth hospital. Hosp Pediatr. 2018;8(5): 102. Moore MR, Schrag SJ, Schuchat A. 112. Saari A, Virta LJ, Sankilampi U, Dunkel
243–250 Effects of intrapartum antimicrobial L, Saxen H. Antibiotic exposure in
93. Berardi A, Fornaciari S, Rossi C, et al. prophylaxis for prevention of group-B- infancy and risk of being overweight in
Safety of physical examination alone for streptococcal disease on the incidence the first 24 months of life. Pediatrics.
managing well-appearing neonates $ and ecology of early-onset neonatal 2015;135(4):617–626
35 weeks’ gestation at risk for early- sepsis. Lancet Infect Dis. 2003;3(4):
113. Roesch LF, Silveira RC, Corso AL, et al.
onset sepsis. J Matern Fetal Neonatal 201–213
Diversity and composition of vaginal
Med. 2015;28(10):1123–1127 103. Green PA, Singh KV, Murray BE, Baker microbiota of pregnant women at risk
94. Joshi NS, Gupta A, Allan JM, et al. CJ. Recurrent group B streptococcal for transmitting Group B Streptococcus
Clinical monitoring of well-appearing infections in infants: clinical and treated with intrapartum penicillin.
infants born to mothers with microbiologic aspects. J Pediatr. 1994; PLoS One. 2017;12(2):e0169916
chorioamnionitis. Pediatrics. 2018; 125(6 pt 1):931–938
114. Aloisio I, Mazzola G, Corvaglia LT, et al.
141(4):e20172056 104. Moylett EH, Fernandez M, Rench MA, Influence of intrapartum antibiotic
95. Joshi NS, Gupta A, Allan JM, et al. Hickman ME, Baker CJ. A 5-year review prophylaxis against group B
Management of chorioamnionitis- of recurrent group B streptococcal Streptococcus on the early newborn
exposed infants in the newborn nursery disease: lessons from twin infants. Clin gut composition and evaluation of the
using a clinical examination-based Infect Dis. 2000;30(2):282–287 anti-Streptococcus activity of
approach. Hosp Pediatr. 2019;9(4): Bifidobacterium strains. Appl Microbiol
105. Fernandez M, Rench MA, Albanyan EA,
227–233 Biotechnol. 2014;98(13):6051–6060
Edwards MS, Baker CJ. Failure of
96. Mukhopadhyay S, Puopolo KM. Clinical rifampin to eradicate group B 115. Azad MB, Konya T, Persaud RR, et al;
and microbiologic characteristics of streptococcal colonization in infants. CHILD Study Investigators. Impact of
early-onset sepsis among very low Pediatr Infect Dis J. 2001;20(4):371–376 maternal intrapartum antibiotics,

16 FROM THE AMERICAN ACADEMY OF PEDIATRICS


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest
method of birth and breastfeeding on prophylaxis upon the intestinal B streptococcus vaccine in healthy
gut microbiota during the first year of microbiota and the prevalence of women and their infants: a randomised
life: a prospective cohort study. BJOG. antibiotic resistance genes in vaginally phase 1b/2 trial. Lancet Infect Dis. 2016;
2016;123(6):983–993 delivered full-term neonates. 16(8):923–934
116. Corvaglia L, Tonti G, Martini S, et al. Microbiome. 2017;5(1):93
122. Hillier SL, Ferrieri P, Edwards MS, et al.
Influence of intrapartum antibiotic 119. Pärnänen K, Karkman A, Hultman J, A phase 2, randomized, control trial of
prophylaxis for group B streptococcus et al. Maternal gut and breast milk group B streptococcus (GBS) type III
on gut microbiota in the first month of microbiota affect infant gut antibiotic capsular polysaccharide-tetanus toxoid
life. J Pediatr Gastroenterol Nutr. 2016; resistome and mobile genetic elements. (GBS III-TT) vaccine to prevent vaginal
62(2):304–308 Nat Commun. 2018;9(1):3891 colonization with GBS III. Clin Infect Dis.
117. Stearns JC, Simioni J, Gunn E, et al. 120. Edwards MS, Baker CJ. Group B 2019;68(12):2079–2086
Intrapartum antibiotics for GBS streptococcal infections in elderly 123. Dzanibe S, Madhi SA. Systematic review
prophylaxis alter colonization patterns adults. Clin Infect Dis. 2005;41(6): of the clinical development of group B
in the early infant gut microbiome of low 839–847 streptococcus serotype-specific
risk infants. Sci Rep. 2017;7(1):16527 121. Madhi SA, Cutland CL, Jose L, et al. capsular polysaccharide-based
118. Nogacka A, Salazar N, Suárez M, et al. Safety and immunogenicity of an vaccines. Expert Rev Vaccines. 2018;
Impact of intrapartum antimicrobial investigational maternal trivalent group 17(7):635–651

PEDIATRICS Volume 144, number 2, August 2019 17


Downloaded from http://publications.aap.org/pediatrics/article-pdf/144/2/e20191881/1077783/peds_20191881.pdf
by guest

You might also like