You are on page 1of 8

Original research

Arch Dis Child Fetal Neonatal Ed: first published as 10.1136/archdischild-2018-316659 on 11 July 2019. Downloaded from http://fn.bmj.com/ on November 2, 2022 at World Health
Effect of intrapartum antibiotics on the intestinal
microbiota of infants: a systematic review
Petra Zimmermann ‍ ‍ ,1,2,3,4 Nigel Curtis2,3,4

1
Department of Paediatrics, Abstract
Fribourg Hospital HFR and Introduction  The use of intrapartum antibiotic What is already known on this topic?
Faculty of Science and Medicine,
University of Fribourg, Fribourg, prophylaxis (IAP) has become common practice in
►► Intrapartum antibiotic prophylaxis (IAP) has
Switzerland obstetric medicine and is used in up to 40% of deliveries.
become common practice in obstetric medicine
2
Department of Paediatrics, Despite its benefits, the risks associated with exposing
The University of Melbourne, and is used in up to 40% of deliveries, which
large numbers of infants to antibiotics, especially
Parkville, Victoria, Australia makes it the most frequent source of antibiotic
3 long-­term effects on health through changes in the
Infectious Diseases Unit, exposure in neonates. IAP is commonly used
The Royal Children’s Hospital microbiota, remain unclear. This systematic review
in women who are colonised with group B
Melbourne, Parkville, Victoria, summarises studies that have investigated the effect of
streptococcus (GBS) and also for caesarean
Australia IAP on the intestinal microbiota of infants.
4
Infectious Diseases Research section surgical prophylaxis.
Methods  A systematic search in Ovid MEDLINE was
Group, Murdoch Children’s ►► The intestinal microbiota plays an important
Research Institute, Parkville, used to identify original studies that investigated the
role in the development of the immune system.
Victoria, Australia effect of IAP on the intestinal microbiota in infants.

Organisation (HINARI) - Group A. Protected by copyright.


There is an early-­life ‘critical window’ during
Studies were excluded if: they included preterm infants,
which the intestinal microbiota and the immune
Correspondence to the antibiotic regimen was not specified, antibiotics were
response develop concurrently. Changes in the
Dr Petra Zimmermann, used for indications other than prophylaxis, probiotics
Department of Paediatrics, composition of the intestinal microbiota in this
were given to mothers or infants, or antibiotics were
Faculty of Science and Medicine, critical period may have a significant influence
University of Fribourg, CH-1700 given to infants. on immune development.
Fribourg, Switzerland; Results  We identified six studies, which investigated
►► The risks associated with exposing a large
p​ etra.z​ immermann@m ​ cri.e​ du.a​ u a total of 272 infants and included 502 stool samples
number of infants to antibiotics, especially
collected up to 3 months of age. In all the studies, IAP
Received 4 December 2018 potential long-­term effects on health through
was given for group B streptococcus (GBS) colonisation.
Revised 29 May 2019 changes in the microbiota, remain unclear.
Accepted 31 May 2019 Infants who were exposed to GBS IAP had a lower
Published Online First bacterial diversity, a lower relative abundance of
11 July 2019 Actinobacteria, especially Bifidobacteriaceae, and a
larger relative abundance of Proteobacteria in their What this study adds?
intestinal microbiota compared with non-­exposed
infants. Conflicting results were reported for the phyla ►► This is the first systematic review summarising
Bacteroidetes and Firmicutes. studies that have investigated the effect of
Conclusions  GBS IAP has profound effects on the IAP on the intestinal microbiota in infants.
intestinal microbiota of infants by diminishing beneficial All eligible studies used IAP for maternal GBS
commensals. Such changes during the early-­life ’critical colonisation.
window’ during which the intestinal microbiota and ►► Infants who were exposed to GBS IAP had a
the immune response develop concurrently may have lower diversity of bacterial species, a lower
an important influence on immune development. The relative abundance of Actinobacteria, especially
potential long-­term adverse consequences of this on the Bifidobacteriaceae, and a larger relative
health of children warrant further investigation. abundance of Proteobacteria in their intestinal
microbiota compared with non-­GBS-­IAP-­
exposed infants.
►► GBS IAP has profound effects on the intestinal
Introduction microbiota of infants by diminishing beneficial
Intrapartum antibiotic prophylaxis (IAP) has commensals. The potential long-­term adverse
become common practice in obstetric medicine and consequences of this on the health of children
is used in 30%–40% of deliveries, which makes warrant further investigation.
it the most frequent source of antibiotic expo-
sure in neonates.1–3 IAP is routinely used in both
elective and emergency caesarean section (CS). deliveries6 7 and GBS colonisation8 are still rising,
© Author(s) (or their
employer(s)) 2020. No In many settings, IAP is also used in women who there is an urgent need to define these effects.9
commercial re-­use. See rights are colonised with group B streptococcus (GBS), The human intestine is the habitat for a large
and permissions. Published resulting in an 80% decrease in the incidence of community of microbes, consisting of archaea,
by BMJ. early-­onset GBS sepsis in infants.4 5 Despite its bacteria, eukaryotes (fungi, helminths and proto-
To cite: Zimmermann P, benefits, the risks associated with exposing such a zoans) and viruses. Microbes are already present
Curtis N. Arch Dis Child Fetal large number of infants to antibiotics, especially its at a low diversity in the placenta and amniotic
Neonatal Ed long-­term effects on health through changes in the fluid,10–12 and the detection of bacteria in meco-
2020;105:F201–F208. microbiota, remain unclear. Since the rates of CS nium suggests that the foetal intestine becomes
Zimmermann P, Curtis N. Arch Dis Child Fetal Neonatal Ed 2020;105:F201–F208. doi:10.1136/archdischild-2018-316659    F201
Original research

Arch Dis Child Fetal Neonatal Ed: first published as 10.1136/archdischild-2018-316659 on 11 July 2019. Downloaded from http://fn.bmj.com/ on November 2, 2022 at World Health
colonised in utero.13–25 After delivery, colonisation of the intes-
tine increases rapidly, and the microbes to which infants are first
exposed play a crucial role in the subsequent establishment of
microbial communities.26 The infant intestine is colonised by
microbes from breast milk, caregivers, family members and the
environment.27 The composition of the intestinal microbiota can
be described on different taxonomic levels (for bacteria, these
comprise phylum, class, order, family, genus and species). The
use of modern sequencing techniques, especially metagenomic
sequencing, allows for detailed analysis down to the species
level. It also enables the detection of antibiotic resistance genes
and the identification of other components of the microbiota
such as fungi and viruses. Another way to analyse the intestinal
microbiota is by measuring bacterial diversity, which entails rich-
ness (number of different bacteria) and evenness (relative abun-
dance of different bacteria). A higher bacterial diversity has been
associated with beneficial effects, for example, stronger vaccine
responses,28 whereas a lower diversity has been associated with
the development of chronic inflammatory bowel disease,29
obesity,30 diabetes mellitus31 and allergic diseases.32 A higher
abundance of Bifidobacterium (belonging to the phylum Acti-
nobacteria) and Lactobacillus (phylum Firmicutes) has also been

Organisation (HINARI) - Group A. Protected by copyright.


associated with beneficial health benefits,33 34 while the role of
other bacteria is less clear. On a species level, a high prevalence
of Bifidobacterium infantis has been associated with increased Figure 1  Selection of studies.
vaccine responses28 and with a reduced risk of infection in
infancy, as well as a reduced risk of chronic illnesses later in
life.35–37 In contrast to bacteria, there is much less known about or infants, or antibiotics were given to infants. References were
the abundance of archaea, eukaryotes or viruses in the intes- hand-­searched for additional publications and no further rele-
tinal microbiota (especially in infants) and their association with vant studies were found. The selection of studies is summarised
health outcomes. Changes in numbers of bacteriophages have in figure 1. The following variables were extracted from the
been associated with chronic inflammatory bowel diseases,38 and included studies: year of study, country, study design, number of
a perturbed intestinal virome has been associated with severe participants, delivery mode, feeding method, antibiotic regimen
malnutrition in infants.39 However, in addition to the abun- (substance, dose, administration route, duration), antibiotic use
dance and diversity of different microbes their metabolites may in pregnancy or in infants, probiotic use, number of collected
also affect health outcomes. For example, bacteria in the colon, stool samples, age at the collection of samples, stool analysis
especially anaerobic bacteria, metabolise undigested carbohy- technique (including sequenced region, sequencing machine,
drates into short-­chain fatty acids (SCFAs). A lack of SCFAs can database used for identification), diversity and relative abun-
disrupt the integrity of colonic epithelial cells, the function of dance of microbiota in stool, changes in bacterial resistance and
the mucosa and the regulation of T cells.40–42 In adults, antibiotic changes in concentrations of SCFAs in stool. Diversity in study
use has been associated with a decrease in SCFAs.43–47 design and reporting precluded quality evaluation according to
While the effect of delivery mode and feeding methods on the the PRISMA (Preferred Reporting Items for Systematic Reviews
establishment of microbial communities in the intestine has been and Meta-­Analyses) guidelines.
well studied, much less is known about the effects of intrapartum
and postpartum antibiotics on the intestinal microbiota.27 In this SYSTEMATIC REVIEW OVERVIEW
review, we systematically review studies that have investigated A total of six studies (all prospective cohort studies) reporting
the effect of IAP on the intestinal microbiota of infants. results from 502 stool samples from 272 infants (127 exposed
to IAP and 145 not exposed to IAP) met the inclusion criteria of
this review (table 1).48–53 All studies were done in west European
SYSTEMATIC REVIEW METHODS countries and all used IAP for GBS colonisation. Studies were
In May 2019, MEDLINE (1946 to present) was searched using rated to be at low risk of bias. None of the studies had any detec-
the Ovid interface with the following search terms: (anti-­ tion, attrition or reporting bias (table 2). However, two studies
bacterial agents OR antibiotic OR prophylaxis OR penicillin OR were deemed to have performance bias because antibiotic use in
ampicillin OR amoxicillin OR amoxycillin OR macrolides OR pregnancy was not reported.51 52 Four studies were done by the
streptococcal OR Streptococcus agalactiae) AND (microbio* same research group in the same time frame, so they have made
OR feces OR faeces OR gastrointestinal microbiome OR culture a potential selection bias, as it is unclear if there was an overlap
OR polymerase chain reaction OR RNA, ribosomal, 16S OR of participants.49–52
high-­throughput nucleotide sequencing) AND (perinatal OR The number of infants in each study ranged from 20 to 84
intrapartum OR pregnancy OR delivery OR labor OR labour (median 45, mean 45). The longest follow-­up (time at the collec-
OR obstetric OR maternal OR neonat* OR newborn OR infant) tion of last stool sample) was 3 months.48 Antibiotic regimens
without any language limitations. This identified 328 articles. were intravenous ampicillin (n=4),49–52 amoxicillin (n=1)53 and
Studies were excluded if: they included preterm infants, the anti- penicillin (n=1).48 Seventy-­four per cent of infants (200 of 272)
biotic regimen was not specified, antibiotics were used for indi- were exclusively breast fed at the time of stool collection. Only
cations other than prophylaxis, probiotics were given to mothers one child was born by CS.53
F202 Zimmermann P, Curtis N. Arch Dis Child Fetal Neonatal Ed 2020;105:F201–F208. doi:10.1136/archdischild-2018-316659
Table 1  Summary of the main findings of studies investigating the effect of GBS IAP on the infant intestinal microbiota
No of:
Author Infants, samples
Country IAP, no-­IAP Analysis techniques Higher abundance in infants with IAP Lower abundance in infants with IAP
Year Characteristics of participants (including region, (relative abundance IAP, relative abundance (relative abundance IAP, relative abundance
Study design Antibiotic regimen machine, database) Age no-­IAP) no-­IAP) Other findings in infants with IAP
Nogacka et al48 40, 160 16S rRNA 2 days Phylum level Lower alpha diversity at all time points (numbers of
Spain 18, 22 V3–V4 Verrucomicrobia (BF-­IAP 0.16, BF-­no-­IAP 0.04, OTUs, Chao1 index and Shannon index)
2017 CS: – MiSeq Illumina p<0.05) Higher number of beta-­lactamase coding genes at
PCS BF: IAP 11/18, no-­IAP 18/22 (3 months) SILVA database Family level all time points
Penicillin intravenous 5 million units followed by Muribaculaceae (0.43, 0.04, p<0.01) Lower propionate levels at 2 days
2.5 million units every 4 hours until delivery PCR for resistance genes Prevotellaceae (0.12, 0.02, p=0.04) Lower levels of propionate at 2 days, acetate
No antibiotics in pregnancy Rikenellaceae (0.28, 0.12, p=0.02) at 10 days and butyrate at 1 and 3 months
No antibiotics given to infants 10 days Phylum level Phylum level Higher levels of propionate at 10 days, 1 month
Bacteroidetes (FF-­IAP 11.8, FF-­no-­IAP 0.32, p<0.05) Actinobacteria (BF-­IAP 12.30, BF-­no-­IAP 22.66, and 3 months and butyrate at 10 days
Firmicutes (BF-­IAP 45.29, BF-­no-­IAP 21.65, p<0.05) p<0.05, FF-­IAP 6.06, FF-­no-­IAP 34.01, p<0.05)
Family level Bacteroidetes (BF-­IAP 1.66, BF-­no-­IAP 22.03,
Muribaculaceae (0.24, 0.04, p=0.05) p<0.05)
Clostridiaceae (16.04, 0.59, p=0.03) Family level
Bifidobacteriaceae (8.87, 22.14, p=0.01)
1 month Family level
Muribaculaceae (0.46, 0.22, p=0.04)
3 months Phylum level
Firmicutes (BF-­IAP 40.41, BF-­C 22.23, p<0.05)
Family level
Campylobacteraceae (0.03, 0.00, p=0.04)
Helicobacteraceae (0.87, 0.03, p=0.04)
Muribaculaceae (0.25, 0.03, p=0.03)
Prevotellaceae (0.08, 0.03, p=0.03)
Mazzola et al49 26, 52 16S rRNA 7 days Family level Phylum level Lower alpha diversity at 7 days (numbers of
Italy 13, 13 V3–V4 Enterobacteriaceae (BF-­IAP 52, BF-­no-­IAP 14, Actinobacteria (BF-­IAP 0, BF-­no-­IAP 17%, p≤0.01) OTUs, Chao1 index and Shannon index)
2016 CS: – MiSeq Illumina p=0.04, FF-­IAP 52, FF-­no-­IAP 15, p=0.13) Genus level
PCS BF: IAP 7/13, no-­IAP 7/13 (1 month) SILVA database Genus level Bifidobacterium (BF-­IAP 0, BF-­no-­IAP 16, p<0.01)
Ampicillin intravenous 2 g, followed by 1 g every Escherichia  (BF-­IAP 52, BF-­no-­IAP 14, p=0.04, MF-­
4 hours until delivery qPCR (Bifidobacterium) IAP 52, MF-­no-­IAP 15, p=0.13)
No antibiotics or probiotics given to infants 1 month Family level Genus level
Veillonellaceae (n.s., p=0.04) Streptococcus (n.s., p=0.04)
Corvaglia et al50 84, 168 qPCR 7 days Genus level No difference in aubundance of Bifidobacterium

Zimmermann P, Curtis N. Arch Dis Child Fetal Neonatal Ed 2020;105:F201–F208. doi:10.1136/archdischild-2018-316659


Italy 35, 49 Bacteroides, ­ Bifidobacterium (6.61, 7.80, p<0.01) at 1 month
2016 CS: – Bifidobacterium, ­ No differences in abundance of Bacteroides or
PCS BF: IAP 26/35, no-­IAP 30/49 (7 days), IAP 22/35, no-­ Lactobacillus ­ Lactobacillus at both time points
IAP 29/49 (1 month) ­
Ampicillin intravenous 2 g, followed by 1 g every ­
4 hours until delivery ­
No antibiotics given to mothers in last 4 weeks of 1 month
pregnancy
No antibiotics or probiotics given to infants
Aloisio et al51 20, 20 16S rRNA 6–7 days Phylum level Phylum level Lower alpha diversity (numbers of OTUs, Chao1
Italy 10, 10 V2–V4, V6–V9 Proteobacteria (54.7, 15.5, p<0.05) Actinobacteria (0.4, 3.8, p<0.05) index and Shannon index)
2016 CS: – Ion Torrent Personal Bacteroidetes (16, 47.8, p<0.05) Higher abundance of Gram-­negative bacteria
PCS BF: 20/20 (7 days) Genome Machine Genus level
Ampicillin intravenous 2 g, followed by 1 g every Ion Reporter Software Bifidobacterium (0.00, 0.07, p<0.05)
4 hours until delivery
Antibiotic use in pregnancy not reported
No antibiotics or probiotics given to infants

Continued
Original research

F203
Organisation (HINARI) - Group A. Protected by copyright.
Arch Dis Child Fetal Neonatal Ed: first published as 10.1136/archdischild-2018-316659 on 11 July 2019. Downloaded from http://fn.bmj.com/ on November 2, 2022 at World Health
Original research

Arch Dis Child Fetal Neonatal Ed: first published as 10.1136/archdischild-2018-316659 on 11 July 2019. Downloaded from http://fn.bmj.com/ on November 2, 2022 at World Health
Multiple methods were used to determine the bacterial intes-

No difference in number of beta-­lactamase resistant


Bifidobacterium, Enterobacteria., Enterococcus, and

16S rRNA, 16S rRNA gene sequencing; BF, exclusively breast fed; CS, caesarean section; DGGE, denaturing gradient gel electrophoresis; FF, partly or exclusively formula fed; IAP, intrapartum antibiotic prophylaxis; n.s., not specified; OTU, operational taxonomic unit; PCS, prospective cohort study; qPCR,
tinal microbiota, including bacterial culture (n=1),53denaturing

B. breve, B. bifidum and B.dentium were most


gradient gel electrophoresis (DGGE) (n=1),52 quantitative PCR

No difference in abundance of Bacteroides,


(n=3),49 50 52 16S rRNA gene sequencing (n=3) (Illumina MiSeq

Lower diversity of Bifidobacterium spp.


Other findings in infants with IAP
(n=2)48 49 and Ion Torrent Personal Genome Machine (n=1).51

No differences of other bacteria


Bacterial diversity was assessed in three of the six studies.48 49
Results were reported down to the lowest taxonomic level anal-
ysed in each study.

Staphylococcus,
Systematic review results
affected

bacteria
Diversity
All three studies that investigated alpha diversity (a measure
of intrasample diversity) of the intestinal microbiota reported
Clostridium (infants colonised 12%, 40%, p<0.05)
(relative abundance IAP, relative abundance

significantly lower diversity in infants who were exposed


to GBS IAP compared with those who were not.48 49 51 The
Lower abundance in infants with IAP

observed numbers of operational taxonomic units (a measure


Bifidobacterium (5.85, 7.29, p<0.01)

of richness through observed taxa without correction for non-­


observed taxa), the Chao1 index (a measure of estimated rich-
ness corrected for relative uncertainty of predicting taxa) and
the Shannon index (a measure of estimated richness and even-
ness corrected for relative uncertainty of predicting taxa) were
Genus level

Genus level

significantly lower in GBS IAP-­exposed infants at a number of

Organisation (HINARI) - Group A. Protected by copyright.


no-­IAP)

time points between 2 days and 3 months of age.48 49 51

Differences at the phylum level


(relative abundance IAP, relative abundance

Three studies reported statistically significant differences in rela-


tive bacterial abundance at the phylum level.48 49 52 In one study,
Higher abundance in infants with IAP

a higher relative abundance of Verrucomicrobia was reported


in infants at 2 days of age who were exclusively breast fed and
exposed to GBS IAP (BF-­GBS IAP).48 In all three studies, infants
exposed to GBS IAP, independent of breast feeding, had a lower
relative abundance of Actinobacteria at 7 to 10 days of age.48 49 52
At the same age, BF-­GBS IAP infants were reported to have a
lower relative abundance of Bacteroidetes than BF-­no-­GBS IAP
no-­IAP)

infants.48 52 In contrast, a higher relative abundance of Bacte-


roidetes was observed in a small number of infants (n=11) who
were partly or exclusively formula fed and exposed to GBS IAP
6–7 days

(FF-­GBS IAP) in one study.48 Additionally, BF-­GBS IAP infants


3 days
Age

were reported to have a higher relative of Proteobacteria at


7 days52 and Firmicutes at 10 days and 3 months of age than
B. fragilis, Bifidobacterium,

BF-­no-­GBS IAP infants.48


Analysis techniques

machine, database)
(including region,

Differences at the family level


Lactobacillus
C. difficile,

Only two of the six studies reported differences in relative bacte-


Culture
E. coli,

DGGE

rial abundance at the family level.48 49 A higher relative abun-


qPCR

dance of Muribaculaceae was reported in infants exposed to GBS


IAP at all measured time points (2, 10 days, 1 and 3 months)
Amoxicillin intravenous 2 g, followed by 1 g every

No antibiotics given to mothers in last 15 days of


Ampicillin intravenous 2 g, followed by 1 g every

in one study.48 Additionally, infants exposed to GBS IAP were


No antibiotics or probiotics given to infants

reported to have a higher relative abundance of Prevotellaceae at


Antibiotic use in pregnancy not reported

7 days and 3 months,48Rikenellaceae at 2 days,48 Enterobacteri-


BF: IAP 26/26, no-­IAP 26/26 (7 days)

aceae at 7 days,49 Clostridiaceae at 10 days,48 Veillonellaceae at


Characteristics of participants

1 month49 and Campylobacteraceae and Helicobacteraceae at 3


months of age.48 In contrast, infants exposed to GBS IAP were
CS: IAP 1/25, no-­IAP 1/25
4 hours until delivery

4 hours until delivery


Antibiotic regimen

reported to have a lower relative abundance of Bifidobacteria-


BF: 34/50 (3 days)
Infants, samples

ceae at the age of 10 days.48


quantitative polymerase chain reaction.
IAP, no-­IAP

pregnancy
No of:

52, 52
26, 26

50, 50
25, 25
Table 1  Continued

Differences at the genus level


CS: –

All, but one study reported differences in the relative abundance


of bacteria at the genus level.49 50 52–54At 7 days of age, a higher
Jauréguy et al53

relative abundance of Escherichia and a lower relative abun-


Study design
Aloisio et al52

dance of Bifidobacterium were reported in infants exposed to


Country
Author

GBS IAP.49 In the same group, a lower abundance of Clostridium


France
2014

2004
Year

Italy

PCS

PCS

(proportion of infants colonised) at the age of 3 days53 and a


F204 Zimmermann P, Curtis N. Arch Dis Child Fetal Neonatal Ed 2020;105:F201–F208. doi:10.1136/archdischild-2018-316659
Original research

Arch Dis Child Fetal Neonatal Ed: first published as 10.1136/archdischild-2018-316659 on 11 July 2019. Downloaded from http://fn.bmj.com/ on November 2, 2022 at World Health
Table 2  Risk of bias summary of studies included in the review
Reference Publication year Selection bias Performance bias Detection bias Attrition bias Reporting bias
Nogacka et al48 2017 − − − − −
Mazzola et al49 2016 +* − − − −
Corvaglia et al50 2016 +* − − − −
Aloisio et al51 2016 +* + − − −
Aloisio et al52 2014 +* + − − −
Jauréguy et al53 2004 − − − − −
*Potential overlap in participants.

lower relative abundance of Bifidobacterium at 7 days50  52 and increased risk of developing allergic diseases and asthma later
Streptococcus at 1 month of age were reported.49 In contrast, in life.65–68
a higher relative abundance of Bacteroides and Parabacteroides This review shows that GBS IAP has profound effects on the
was reported at 3 months of age.54 intestinal microbiota in infants (summarised in figure 2). Infants
who are exposed to GBS IAP have a lower bacterial diversity of
Differences at the species level their bacterial intestinal microbiota, a lower relative abundance
One study used DGGE to differentiate which Bifidobacterium of Actinobacteria, especially Bifidobacteriaceae, and a larger
spp. were most affected by GBS IAP and showed that the species relative abundance of Proteobacteria compared with infants who
which were most depleted in IAP-­exposed infants were B. breve, are not exposed to GBS IAP. Similarly, a study which was not
B. bifidum and B. dentium.52 Additionally, these infants also had included in this review because some of the infants were treated

Organisation (HINARI) - Group A. Protected by copyright.


a lower diversity of Bifidobacterium spp.52 with antibiotics after delivery also reported a delay in expan-
sion of Bifidobacteriumduring the first 12 weeks of life in infants
Differences in bacterial antibiotic resistance whose mothers received GBS IAP. This study also reported a
A recent study used PCR to identify and quantify resistance persistence of Escherichia in these infants.69 Furthermore, it
genes.48 It reported that infants exposed to GBS IAP have a is likely that GBS IAP also reduces the numbers of Bacteroi-
higher numbers of beta-­lactamase coding genes at all four time detes as reported in two studies in this review.48 52 Additionally,
points tested (2 and 7 days, 1 and 3 months), but no increase in another study which was also not included in this review because
genes coding for resistance to tetracyclines.48 The oldest study it included infants who were given antibiotics after birth also
included in the review, which used culture for the identification reported a significantly lower relative abundance of Bacteroi-
of bacteria, did not observe any effect of GBS IAP exposure on detes at the age 3 and 12 months and an over-­representation
the proportion of beta-­lactamase resistant bacteria.53 of Enterococcus and Clostridium in infants exposed to IAP.54
Although one study reported the opposite (a higher relative
abundance of Bacteroidetes in FF-­ GBS IAP-­ exposed infants),
Differences in faecal short-chain fatty acids
it only included 11 samples.48 Studies investigating the relative
One study compared faecal SCFAs between GBS IAP-­exposed
abundance of Firmicutes report conflicting results. This might
and non-­exposed infants. The former had significantly lower
be attributable to heterogeneity in study design, stool analysis
levels of propionate at 2 days, acetate at 10 days and butyrate
techniques, as well as the DNA isolation, amplification and
at 1 and 3 months of age. In contrast, GBS IAP-­exposed infants 
sequencing protocols in those studies which used sequencing.
had significantly higher propionate levels at 10 days, 1 and 3
Only one study reported changes in Verrucomicrobia, which is
months of age and higher butyrate levels at 10 days of age.48

Discussion
The intestinal microbiota plays an important role in the devel-
opment of the immune system. There is an early-­life ‘critical
window’ during which the intestinal microbiota and the immune
response develop concurrently. The development of intestinal
microbiota is easily disrupted by external factors,27 and pertur-
bation of the intestinal microbiota during this vulnerable period
may have a large influence on immune development.21 55–59
Numerous studies suggest that the composition of intestinal
microbiota is associated with many immune- mediated and non-­
immune-­ mediated diseases, including sepsis18 and necrotising
enterocolitis (NEC) in neonates.60 It is suggested that the intes-
tinal microbiota also play an important role in the development
of chronic inflammatory bowel disease,29 diabetes mellitus31 and
allergic diseases in children.34
IAP has been associated with adverse clinical outcomes. For
example, administration of amoxicillin/clavulanate as IAP
leads to increased rates of NEC in preterm infants.61 IAP has
also been associated with amoxicillin-­resistant late-­onset E. coli
infections.62 63 Furthermore, antibiotic exposure in the first 6 Figure 2  Summary of the main differences in the intestinal microbiota
months of life has been associated with long-­term adverse health between infants who were or were not exposed to GBS IAP (reported at
outcomes, such as a 20% increased risk of obesity64 and an the lowest taxonomic level that was analysed in each study).
Zimmermann P, Curtis N. Arch Dis Child Fetal Neonatal Ed 2020;105:F201–F208. doi:10.1136/archdischild-2018-316659 F205
Original research

Arch Dis Child Fetal Neonatal Ed: first published as 10.1136/archdischild-2018-316659 on 11 July 2019. Downloaded from http://fn.bmj.com/ on November 2, 2022 at World Health
likely because of its very low abundance in the first few weeks that many of the studies were limited by small sample size, short
of life. follow-­up and inadequate accounting for potential confounding
Unfortunately, the studies included in this review followed up factors. A further limitation is that four of the six studies were
infants only until the age of 3 months. The above-­mentioned done by the same research group, and there was potentially an
study, which was not included, showed that IAP-­induced changes overlap of participants.49–52 However, different stool analysis
in the intestinal microbiota were still evident at the age of 12 techniques were used in these studies (PCR50 52 and 16S rRNA
months in infants who were born by emergency CS, but not in gene sequencing49 51).
infants exposed to IAP for other reasons.54 However, since the Further studies are needed to clarify whether restricted use of
first few weeks of life are important in training mucosal immu- IAP has adverse outcomes for neonates. The benefits of IAP need
nity, the effect of IAP might have profound effects on health in to be weighed against the potential (especially long-­term) adverse
later childhood. effects. This will require larger studies with longer follow-­up
A likely explanation for the observed patterns of differences to investigate the effect of IAP on the intestinal microbiota of
in the intestinal microbiota induced by GBS IAP is that peni- infants and relate changes with clinical outcomes later in life.
cillin and ampicillin have stronger activity on Gram-­ positive Once the relationship between the intestinal microbiota and the
bacteria, allowing overgrowth of Gram-­negative bacteria such as development of the immune system is clearer, interventions such
Proteobacteria. GBS IAP leads to a decrease in bacteria which are as exclusive breast feeding, targeted probiotic administration
considered beneficial commensals, such as Bifidobacteriaceae, or phage therapy might be used as adjuvant therapy in infants
but to an increase in potentially pathogenic bacteria, including exposed to antibiotics. Furthermore, the successful development
Enterobacteriaceae,49 Campylobacteriaceae and Helicobacteri- of a GBS vaccine would help reduce the necessity for IAP.
aceae.48 These changes (higher rates of Enterobacteriaceae and Correction notice  The article type has been changed to Original article since this
lower rates of Bifidobacteriaceae), which are also found in chil- paper was published Online First.
dren born by CS, have been associated with an increased risk of Acknowledgements  PZ greatly acknowledges funding recieved from the

Organisation (HINARI) - Group A. Protected by copyright.


developing allergic diseases and asthma.34 European Society of Paediatric Infectious Diseases (ESPID) and the University of
Additionally, a higher relative abundance of the phylum Melbourne.
Proteobacteria has been associated with lower vaccine responses, Contributors  PZ drafted the initial manuscript. NC critically revised the manuscript
while a higher relative abundance of the phylum Actinobacteria and both authors approved the final manuscript as submitted.
has been associated with higher vaccine responses.33 Both these Funding  PZ is supported by a Fellowship from EPSID and an International Research
changes (higher relative abundance of Proteobacteria48 52 and Scholarship from the University of Melbourne.
lower abundance of Actinobacteria)48 49 52 have been associated Competing interests  None declared.
with IAP. Furthermore, Bifidobacteriaceae, which are suppressed Patient consent for publication  Not required.
through antibiotics have been associated with beneficial health Provenance and peer review  Not commissioned; externally peer reviewed.
outcomes, such as inhibiting the growth of potentially patho-
genic bacteria and preventing intestinal infection70 71 and allevi- ORCID iD
ation of constipation.72 73 Petra Zimmermann http://​orcid.​org/​0000-​0002-​2388-​4318
Some of the studies included in this review reported a larger
effect of IAP on the intestinal microbiota in infants who are References
exclusively breast fed,49 while others found less changes in 1 Van Dyke MK, Phares CR, Lynfield R, et al. Evaluation of universal antenatal screening
breastfed infants born by CS.54 for group B streptococcus. N Engl J Med 2009;360:2626–36.
2 Persaud RR, Azad MB, Chari RS, et al. Perinatal antibiotic exposure of neonates
As only one study investigated the effect of GBS IAP on
in Canada and associated risk factors: a population-­based study. J Matern Fetal
SCFA (reporting lower butyrate and higher propionate levels) Neonatal Med 2015;28:1190–5.
precludes drawing any firm conclusion on the effect of GBS IAP 3 Stokholm J, Schjørring S, Pedersen L, et al. Prevalence and predictors of antibiotic
on SCFA.48 Higher butyrate and propionate levels in early life administration during pregnancy and birth. PLoS One 2013;8:e82932.
have been associated with protection against developing allergic 4 Jordan HT, Farley MM, Craig A, et al. Revisiting the need for vaccine prevention of
late-­onset neonatal group B streptococcal disease: a multistate, population-­based
diseases later in life.74 Butyrate and propionate have also been analysis. Pediatr Infect Dis J 2008;27:1057–64.
shown to protect against obesity.75 5 Active Bacterial Core Surveillance Report, Emerging Infections Program Network,
The use of IAP is rising as a result of increasing rates of Group B Streptococcus. 2014 (Accessed Feb 2017).
both CS and GBS colonisation. In the USA alone, GBS IAP 6 Mueller M, Kolly L, Bauman M, et al. Analysis of caesarean section rates over time
in a single Swiss centre using a ten-­group classification system. Swiss Med Wkly
leads to approximately 1 million infants exposed to antibiotics
2014;144:w13921.
each year.76 There is still debate about whether universal GBS 7 Jayot A, Nizard J. Evolution of cesarean categories in a modified Robson classification
screening and routine administration of antibiotics to colonised in a single center from 2002 to 2012 due to high rate of maternal pathology. J Obstet
mothers leads to better outcomes than risk-­based screening.77–79 Gynaecol Res 2016;42:648–54.
Furthermore, IAP is often used in situations in which it has not 8 Stapleton RD, Kahn JM, Evans LE, et al. Risk factors for group B streptococcal
genitourinary tract colonization in pregnant women. Obstet Gynecol
been proven to have a clear benefit, such as prelabour rupture of 2005;106:1246–52.
membranes in term deliveries80 and preterm labour with intact 9 Sandall J, Tribe RM, Avery L, et al. Short-­term and long-­term effects of caesarean
membranes.81 A minority of countries, such as Norway, do not section on the health of women and children. Lancet 2018;392:1349–57.
use IAP for elective CS, without adverse outcomes.82 None of the 10 Collado MC, Rautava S, Aakko J, et al. Human gut colonisation may be initiated in
utero by distinct microbial communities in the placenta and amniotic fluid. Sci Rep
studies included in this review commented on adverse outcomes
2016;6:23129.
of infants whose mothers did not receive IAP. 11 Stout MJ, Conlon B, Landeau M, et al. Identification of intracellular bacteria in the
The strengths of this review are that the included studies used basal plate of the human placenta in term and preterm gestations. Am J Obstet
similar antibiotic regimens (intravenous ampicillin or penicillin), Gynecol 2013;208:226.e1–7.
the homogeneity of infants included in the studies (mostly vagi- 12 Aagaard K, Ma J, Antony KM, et al. The placenta harbors a unique microbiome. Sci
Transl Med 2014;6:237ra65.
nally born, exclusively breastfed infants) and the use of advanced 13 Martin R, Makino H, Cetinyurek Yavuz A, et al. Early-­life events, including mode
DNA sequencing techniques in most studies. The main limitation of delivery and type of feeding, siblings and gender, shape the developing gut
of this review, apart from the relatively low number of studies, is microbiota. PLoS One 2016;11:e0158498.

F206 Zimmermann P, Curtis N. Arch Dis Child Fetal Neonatal Ed 2020;105:F201–F208. doi:10.1136/archdischild-2018-316659
Original research

Arch Dis Child Fetal Neonatal Ed: first published as 10.1136/archdischild-2018-316659 on 11 July 2019. Downloaded from http://fn.bmj.com/ on November 2, 2022 at World Health
14 Jiménez E, Marín ML, Martín R, et al. Is meconium from healthy newborns actually 44 Heimdahl A, Nord CE. Influence of erythromycin on the normal human flora and
sterile? Res Microbiol 2008;159:187–93. colonization of the oral cavity, throat and colon. Scand J Infect Dis 1982;14:49–56.
15 Moles L, Gómez M, Heilig H, et al. Bacterial diversity in meconium of preterm 45 Orrhage K, Brismar B, Nord CE. Effect of Supplements with Bifidobacterium longum
neonates and evolution of their fecal microbiota during the first month of life. PLoS and Lactobacillus acidophilus on the Intestinal Microbiota during Administration of
One 2013;8:e66986. Clindamycin. Microbial Ecology in Health and Disease 1994;7:17–25.
16 Sakata H, Yoshioka H, Fujita K. Development of the intestinal flora in very low 46 Heimdahl A, Nord CE. Effect of phenoxymethylpenicillin and clindamycin on the oral,
birth weight infants compared to normal full-­term newborns. Eur J Pediatr throat and faecal microflora of man. Scand J Infect Dis 1979;11:233–42.
1985;144:186–90. 47 Heimdahl A, Nord CE. Influence of doxycycline on the normal human flora and
17 Ardissone AN, de la Cruz DM, Davis-­Richardson AG, et al. Meconium microbiome colonization of the oral cavity and colon. Scand J Infect Dis 1983;15:293–302.
analysis identifies bacteria correlated with premature birth. PLoS One 48 Nogacka A, Salazar N, Suárez M, et al. Impact of intrapartum antimicrobial
2014;9:e90784. prophylaxis upon the intestinal microbiota and the prevalence of antibiotic resistance
18 Madan JC, Salari RC, Saxena D, et al. Gut microbial colonisation in premature genes in vaginally delivered full-­term neonates. Microbiome 2017;5:93.
neonates predicts neonatal sepsis. Arch Dis Child Fetal Neonatal Ed 49 Mazzola G, Murphy K, Ross RP, et al. Early gut microbiota perturbations following
2012;97:F456–62. intrapartum antibiotic prophylaxis to prevent group B streptococcal disease. PLoS One
19 Rotimi VO, Olowe SA, Ahmed I. The development of bacterial flora of premature 2016;11:e0157527.
neonates. J Hyg 1985;94:309–18. 50 Corvaglia L, Tonti G, Martini S, et al. Influence of intrapartum antibiotic prophylaxis
20 Wampach L, Heintz-­Buschart A, Hogan A, et al. Colonization and succession within for group B streptococcus on gut microbiota in the first month of life. J Pediatr
the human gut microbiome by archaea, bacteria, and microeukaryotes during the first Gastroenterol Nutr 2016;62:304–8.
year of life. Front Microbiol 2017;8:738. 51 Aloisio I, Quagliariello A, De Fanti S, et al. Evaluation of the effects of intrapartum
21 Gosalbes MJ, Llop S, Vallès Y, et al. Meconium microbiota types dominated by lactic antibiotic prophylaxis on newborn intestinal microbiota using a sequencing approach
acid or enteric bacteria are differentially associated with maternal eczema and targeted to multi hypervariable 16S rDNA regions. Appl Microbiol Biotechnol
respiratory problems in infants. Clin Exp Allergy 2013;43:198–211. 2016;100:5537–46.
22 Hansen R, Scott KP, Khan S, et al. First-­Pass meconium samples from healthy 52 Aloisio I, Mazzola G, Corvaglia LT, et al. Influence of intrapartum antibiotic prophylaxis
term vaginally-­delivered neonates: an analysis of the microbiota. PLoS One against group B Streptococcus on the early newborn gut composition and evaluation
2015;10:e0133320. of the anti-­Streptococcus activity of Bifidobacterium strains. Appl Microbiol Biotechnol
23 Makino H, Kushiro A, Ishikawa E, et al. Mother-­to-­infant transmission of intestinal 2014;98:6051–60.
bifidobacterial strains has an impact on the early development of vaginally delivered 53 Jauréguy F, Carton M, Panel P, et al. Effects of intrapartum penicillin prophylaxis on

Organisation (HINARI) - Group A. Protected by copyright.


infant’s microbiota. PLoS One 2013;8:e78331. intestinal bacterial colonization in infants. J Clin Microbiol 2004;42:5184–8.
24 Makino H, Kushiro A, Ishikawa E, et al. Transmission of intestinal Bifidobacterium 54 Azad MB, Konya T, Persaud RR, et al. Impact of maternal intrapartum antibiotics,
longum subsp. longum strains from mother to infant, determined by multilocus method of birth and breastfeeding on gut microbiota during the first year of life: a
sequencing typing and amplified fragment length polymorphism. Appl Environ prospective cohort study. BJOG 2016;123:983–93.
Microbiol 2011;77:6788–93. 55 Ismail IH, Oppedisano F, Joseph SJ, et al. Reduced gut microbial diversity in early life
25 Nagpal R, Tsuji H, Takahashi T, et al. Sensitive quantitative analysis of the meconium is associated with later development of eczema but not atopy in high-­risk infants.
bacterial microbiota in healthy term infants born vaginally or by cesarean section. Pediatr Allergy Immunol 2012;23:674–81.
Front Microbiol 1997;2016:7. 56 Abrahamsson TR, Jakobsson HE, Andersson AF, et al. Low diversity of the gut
26 Tamburini S, Shen N, Wu HC, et al. The microbiome in early life: implications for health microbiota in infants with atopic eczema. J Allergy Clin Immunol 2012;129:434–40.
outcomes. Nat Med 2016;22:713–22. 57 Storrø O, Øien T, Langsrud Ø, et al. Temporal variations in early gut microbial
27 Zimmermann P, Curtis N. Factors Influencing the Intestinal Microbiome During the colonization are associated with allergen-­specific immunoglobulin E but not atopic
First Year of Life. Pediatr Infect Dis J 2018;37:e315–35. eczema at 2 years of age. Clin Exp Allergy 2011;41:1545–54.
28 Huda MN, Lewis Z, Kalanetra KM, et al. Stool microbiota and vaccine responses of 58 Wang M, Karlsson C, Olsson C, et al. Reduced diversity in the early fecal microbiota of
infants. Pediatrics 2014;134:e362–72. infants with atopic eczema. J Allergy Clin Immunol 2008;121:129–34.
29 Manichanh C, Rigottier-­Gois L, Bonnaud E, et al. Reduced diversity of faecal 59 Adlerberth I, Strachan DP, Matricardi PM, et al. Gut microbiota and development of
microbiota in Crohn’s disease revealed by a metagenomic approach. Gut atopic eczema in 3 European birth cohorts. J Allergy Clin Immunol 2007;120:343–50.
2006;55:205–11. 60 Warner BB, Deych E, Zhou Y, et al. Gut bacteria dysbiosis and necrotising
30 Turnbaugh PJ, Hamady M, Yatsunenko T, et al. A core gut microbiome in obese and enterocolitis in very low birthweight infants: a prospective case-­control study. Lancet
lean twins. Nature 2009;457:480–4. 2016;387:1928–36.
31 Knip M, Siljander H. The role of the intestinal microbiota in type 1 diabetes mellitus. 61 Kenyon S, Boulvain M, Neilson JP. Antibiotics for preterm rupture of membranes.
Nat Rev Endocrinol 2016;12:154–67. Cochrane Database Syst Rev 2013:Cd001058.
32 Azad MB, Konya T, Guttman DS, et al. Infant gut microbiota and food sensitization: 62 Didier C, Streicher MP, Chognot D, et al. Late-­onset neonatal infections: incidences
associations in the first year of life. Clin Exp Allergy 2015;45:632–43. and pathogens in the era of antenatal antibiotics. Eur J Pediatr 2012;171:681–7.
33 Zimmermann P, Curtis N. The influence of the intestinal microbiome on vaccine 63 Hyde TB, Hilger TM, Reingold A, et al. Trends in incidence and antimicrobial resistance
responses. Vaccine 2018;36:4433–9. of early-­onset sepsis: population-­based surveillance in San Francisco and Atlanta.
34 Zimmermann P, Messina N, Mohn WW, et al. Association between the intestinal Pediatrics 2002;110:690–5.
microbiota and allergic sensitization, eczema, and asthma: A systematic review. 64 Rasmussen SH, Shrestha S, Bjerregaard LG, et al. Antibiotic exposure in early life and
J Allergy Clin Immunol 2019;143. childhood overweight and obesity: A systematic review and meta-­analysis. Diabetes
35 Sela DA, Mills DA. The marriage of nutrigenomics with the microbiome: the case of Obes Metab 2018;20:1508–14.
infant-­associated bifidobacteria and milk. Am J Clin Nutr 2014;99:697S–703. 65 Droste JH, Wieringa MH, Weyler JJ, et al. Does the use of antibiotics in early childhood
36 Sela DA, Chapman J, Adeuya A, et al. The genome sequence of Bifidobacterium increase the risk of asthma and allergic disease? Clin Exp Allergy 2000;30:1548–53.
longum subsp. infantis reveals adaptations for milk utilization within the infant 66 Johnson CC, Ownby DR. Allergies and asthma: do atopic disorders result from
microbiome. Proc Natl Acad Sci U S A 2008;105:18964–9. inadequate immune homeostasis arising from infant gut dysbiosis? Expert Rev Clin
37 LoCascio RG, Desai P, Sela DA, et al. Broad conservation of milk utilization genes Immunol 2016;12:379–88.
in Bifidobacterium longum subsp. infantis as revealed by comparative genomic 67 Strömberg Celind F, Wennergren G, Vasileiadou S, et al. Antibiotics in the first
hybridization. Appl Environ Microbiol 2010;76:7373–81. week of life were associated with atopic asthma at 12 years of age. Acta Paediatr
38 Norman JM, Handley SA, Baldridge MT, et al. Disease-­specific alterations in the enteric 2018;107:1798–804.
virome in inflammatory bowel disease. Cell 2015;160:447–60. 68 Ong MS, Umetsu DT, Mandl KD. Consequences of antibiotics and infections in infancy:
39 Reyes A, Blanton LV, Cao S, et al. Gut DNA viromes of Malawian twins discordant for bugs, drugs, and wheezing. Ann Allergy Asthma Immunol 2014;112:441–5.
severe acute malnutrition. Proc Natl Acad Sci U S A 2015;112:11941–6. 69 Stearns JC, Simioni J, Gunn E, et al. Intrapartum antibiotics for GBS prophylaxis alter
40 Bhaskaran N, Quigley C, Paw C, et al. Role of short chain fatty acids in controlling Tregs colonization patterns in the early infant gut microbiome of low risk infants. Sci Rep
and immunopathology during mucosal infection. Front Microbiol 2018;9:9. 2017;7:16527.
41 Schilderink R, Verseijden C, Seppen J, et al. The SCFA butyrate stimulates the epithelial 70 Castro-­Bravo N, Wells JM, Margolles A, et al. Interactions of surface
production of retinoic acid via inhibition of epithelial HDAC. Am J Physiol Gastrointest exopolysaccharides from bifidobacterium and lactobacillus within the intestinal
Liver Physiol 2016;310:G1138–46. environment. Front Microbiol 2018;9:2426.
42 Topping DL, Clifton PM. Short-­chain fatty acids and human colonic function: roles of 71 Khodaii Z, Ghaderian SMH, Natanzi MM. Probiotic bacteria and their supernatants
resistant starch and nonstarch polysaccharides. Physiol Rev 2001;81:1031–64. protect enterocyte cell lines from enteroinvasive Escherichia coli (EIEC) invasion. Int J
43 Meijer-­Severs GJ, Van Santen E, Meijer BC. Short-­chain fatty acid and organic Mol Cell Med 2017;6:183–9.
acid concentrations in feces of healthy human volunteers and their correlations 72 Dimidi E, Christodoulides S, Scott SM, et al. Mechanisms of action of probiotics
with anaerobe cultural counts during systemic ceftriaxone administration. Scand J and the gastrointestinal microbiota on gut motility and constipation. Adv Nutr
Gastroenterol 1990;25:698–704. 2017;8:484–94.

Zimmermann P, Curtis N. Arch Dis Child Fetal Neonatal Ed 2020;105:F201–F208. doi:10.1136/archdischild-2018-316659 F207
Original research

Arch Dis Child Fetal Neonatal Ed: first published as 10.1136/archdischild-2018-316659 on 11 July 2019. Downloaded from http://fn.bmj.com/ on November 2, 2022 at World Health
73 Ishizuka A, Tomizuka K, Aoki R, et al. Effects of administration of Bifidobacterium 78 Kurz E, Davis D. Routine culture-­based screening versus risk-­based management
animalis subsp. lactis GCL2505 on defecation frequency and bifidobacterial for the prevention of early-­onset group B streptococcus disease in the neonate: a
microbiota composition in humans. J Biosci Bioeng 2012;113:587–91. systematic review. JBI Database System Rev Implement Rep
74 Roduit C, Frei R, Ferstl R, et al. High levels of butyrate and propionate in early life are 2015;13:206–46.
associated with protection against atopy. Allergy 2019;74. 79 Schrag SJ, Zell ER, Lynfield R, et al. A population-­based comparison of strategies
75 Lin HV, Frassetto A, Kowalik EJ, et al. Butyrate and propionate protect against diet-­ to prevent early-­onset group B streptococcal disease in neonates. N Engl J Med
induced obesity and regulate gut hormones via free fatty acid receptor 3-­independent 2002;347:233–9.
mechanisms. PLoS One 2012;7:e35240. 80 Wojcieszek AM, Stock OM, Flenady V. Antibiotics for prelabour rupture of membranes
76 Martin JA, Hamilton BE, Osterman MJ, et al. Births: final data for 2013. National vital at or near term. Cochrane Database Syst Rev 2014:CD001807.
statistics reports: from the Centers for Disease Control and Prevention, National Center for 81 Flenady V, Hawley G, Stock OM, et al. Prophylactic antibiotics for inhibiting preterm
Health Statistics, National Vital Statistics System. Natl Vital Stat Rep 2015;64:1–65. labour with intact membranes. Cochrane Database Syst Rev 2013:CD000246.
77 Homer CS, Scarf V, Catling C, et al. Culture-­based versus risk-­based screening for 82 Opøien HK, Valbø A, Grinde-­Andersen A, et al. Post-­cesarean surgical site infections
the prevention of group B streptococcal disease in newborns: a review of national according to CDC standards: rates and risk factors. A prospective cohort study. Acta
guidelines. Women Birth 2014;27:46–51. Obstet Gynecol Scand 2007;86:1097–102.

Organisation (HINARI) - Group A. Protected by copyright.

F208 Zimmermann P, Curtis N. Arch Dis Child Fetal Neonatal Ed 2020;105:F201–F208. doi:10.1136/archdischild-2018-316659

You might also like