Você está na página 1de 19

Consensus Guidelines

Neonatology 2017;111:107125 Received: June 16, 2016


Accepted after revision: August 8, 2016
DOI: 10.1159/000448985
Published online: September 21, 2016

European Consensus Guidelines on


the Management of Respiratory Distress
Syndrome 2016 Update
David G. Sweet a Virgilio Carnielli b Gorm Greisen c Mikko Hallman d
Eren Ozek e Richard Plavka f Ola Didrik Saugstad g Umberto Simeoni h
Christian P. Speer i Mximo Vento j Gerard H.A. Visser k Henry L. Halliday l
a
Regional Neonatal Unit, Royal Maternity Hospital, Belfast, UK; b Department of Neonatology, University Polytechnic
della Marche, University Hospital Ancona, Ancona, Italy; c Department of Neonatology, Rigshospitalet and University of
Copenhagen, Copenhagen, Denmark; d Department of Pediatrics and Adolescence, Oulu University Hospital,
and PEDEGO Research Unit, Medical Research Center, University of Oulu, Oulu, Finland; e Division of Neonatology,
Department of Pediatrics, Marmara University School of Medicine, Istanbul, Turkey; f Division of Neonatology,
Department of Obstetrics and Gynecology, General Faculty Hospital, and 1st Faculty of Medicine, Charles University,
Prague, Czech Republic; g Department of Pediatric Research, Oslo University Hospital Rikshospitalet, University of
Oslo, Oslo, Norway; h Division of Pediatrics, CHUV, and University of Lausanne, Lausanne, Switzerland; i Department of
Pediatrics, University Childrens Hospital, Wrzburg, Germany; j Neonatal Research Unit, Department of Pediatrics,
Health Research Institute La Fe, University and Polytechnic Hospital La Fe, Valencia, Spain; k Division of Obstetrics,
Department of Obstetrics and Gynecology, University Medical Centre, Utrecht, The Netherlands; l Department of
Child Health, Royal Maternity Hospital, Queens University Belfast, Belfast, UK

Key Words Abstract


Antenatal steroids Continuous positive airway Advances in the management of respiratory distress syn-
pressure Evidence-based practice Hyaline membrane drome (RDS) ensure that clinicians must continue to revise
disease Mechanical ventilation Nutrition Oxygen current practice. We report the third update of the European
supplementation Patent ductus arteriosus Preterm Guidelines for the Management of RDS by a European panel
infant Respiratory distress syndrome Surfactant therapy of expert neonatologists including input from an expert
Thermoregulation perinatal obstetrician based on available literature up to the
beginning of 2016. Optimizing the outcome for babies with
RDS includes consideration of when to use antenatal ste-
roids, and good obstetric practice includes methods of pre-
These updated guidelines contain new evidence from recent Coch- dicting the risk of preterm delivery and also consideration of
rane reviews and the medical literature since 2013. Strength of evi- whether transfer to a perinatal centre is necessary and safe.
dence supporting recommendations has been evaluated using the
GRADE system. The prenatal care section has been expanded and Methods for optimal delivery room management have be-
updated by Prof. Gerard H.A. Visser. There are also new recommenda- come more evidence based, and protocols for lung protec-
tions covering less invasive surfactant administration. tion, including initiation of continuous positive airway pres-
158.232.240.113 - 9/26/2016 8:31:19 PM

2016 S. Karger AG, Basel Dr. David G. Sweet, MD, FRCPCH


Regional Neonatal Unit, Royal Maternity Hospital
HINARI Guatemala

274 Grosvenor Road


Downloaded by:

E-Mail karger@karger.com
Belfast BT12 6BB (UK)
www.karger.com/neo
E-Mail david.sweet@belfasttrust.hscni.net
sure and titration of oxygen, should be implemented from grams are rarely seen today due to early surfactant ther-
soon after birth. Surfactant replacement therapy is a crucial apy and early continuous positive airway pressure
part of the management of RDS, and newer protocols for sur- (CPAP). Definitions based on blood gas analysis and in-
factant administration are aimed at avoiding exposure to spired oxygen concentrations are also increasingly re-
mechanical ventilation, and there is more evidence of differ- dundant as clinicians have moved towards a more prag-
ences among various surfactants in clinical use. Newer meth- matic approach of giving surfactant therapy based on
ods of maintaining babies on non-invasive respiratory sup- clinical assessment of work of breathing and inspired ox-
port have been developed and offer potential for greater ygen requirement very early in the clinical course of the
comfort and less chronic lung disease. As technology for de- disease. Knowing how many babies have genuine RDS is
livering mechanical ventilation improves, the risk of causing therefore difficult. Of the 4,142 babies from Europe for
lung injury should decrease although minimizing the time whom data were submitted to the Vermont Oxford Net-
spent on mechanical ventilation using caffeine and if neces- work during 2015, RDS was coded for about 80% of ba-
sary postnatal steroids are also important considerations. bies born at 28 weeks gestation, increasing to 95% at 24
Protocols for optimizing the general care of infants with RDS weeks gestation [5]. However, recent large clinical trials
are also essential with good temperature control, careful flu- show that when given early CPAP, babies of 2629 weeks
id and nutritional management, maintenance of perfusion gestation can be managed without intubation or surfac-
and judicious use of antibiotics all being important determi- tant about 50% of the time, and the high reported inci-
nants of best outcome. 2016 S. Karger AG, Basel dence may reflect practice where babies are being coded
as having RDS because they were treated with prophylac-
tic or early surfactant.
The aim of the management of RDS is to provide in-
Introduction terventions that maximize survival whilst minimizing po-
tential adverse effects, including the risk of BPD. Many
Respiratory distress syndrome (RDS) remains a sig- strategies and therapies for prevention and treatment of
nificant problem for preterm babies although its manage- RDS are still being tested in clinical trials, and many new
ment has evolved gradually over the years, resulting in studies are incorporated into updated systematic reviews.
improved survival for the smallest infants but with pos- These guidelines update the previous three guidelines af-
sible increasing rates of bronchopulmonary dysplasia ter critical examination of the most up-to-date evidence
(BPD) at least in part due to the reduced use of postnatal available in early 2016. We have employed a similar for-
steroids [1]. Since 2006, a group of neonatologists from mat of summarizing the relevant issues requiring consid-
various European countries have met on a 3-yearly basis eration followed by evidence-based recommendations
to review the most up-to-date literature and to agree on supported by a score using the GRADE system to reflect
consensus recommendations for the optimal manage- the authors views on the strength of evidence supporting
ment of preterm babies with, or at risk of, RDS in order each of the recommendations [6]. Quality of evidence
to try and achieve the best outcomes for neonates in Eu- and strength of recommendations are summarized in ta-
rope. The European Consensus Guidelines for the Man- ble1.
agement of RDS were first published in 2007, have been
updated in 2010 and 2013 and are endorsed by the Euro-
pean Association of Perinatal Medicine [24]. The guide- Prenatal Care
lines have been translated into several languages, includ-
ing Chinese, and although primarily intended for use in There are no generally effective means to improve the
Europe, they contain recommendations that can poten- outcome of infants by preventing the common causes of
tially be used anywhere, provided clinicians have access either spontaneous or elective preterm births. However,
to resources needed to fulfil the standards present in in pregnant women at risk of spontaneous preterm birth,
modern neonatal intensive care units (NICU). due either to previous preterm birth or where a short cer-
Although primarily a disorder of surfactant deficiency vix has been identified on ultrasound examination, use of
resulting in pulmonary insufficiency from soon after progesterone has been associated with clinical benefits to
birth, the classical pattern of RDS has changed as treat- the infant including reduced preterm delivery rates and
ments have evolved over the years. Classical RDS radio- reduced perinatal mortality [7]. However, the findings
graphic appearances of ground glass with air broncho- may not be generally applicable to all modes of adminis-
158.232.240.113 - 9/26/2016 8:31:19 PM

108 Neonatology 2017;111:107125 Sweet etal.


DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
Table 1. Representations of quality of evidence and strength of (RCTs) in which corticosteroids were given in both arms
recommendations of the trial [16]. Given their limited value, only drugs that
are safe for the mother should be considered, i.e. oxytocin
Quality of evidence
High quality A antagonists or Ca channel blockers [17]. Both drugs have
Moderate quality B similar efficacy and perinatal outcome; the former has the
Low quality C fewest maternal side effects.
Very low quality D Prenatal corticosteroids given to mothers with antici-
Strength of recommendation pated preterm delivery improve survival, reduce the risk
Strong recommendation for using intervention 1
Weak recommendation for using intervention 2 of RDS, NEC and intraventricular haemorrhage, and a
single course does not appear to be associated with any
significant maternal or short-term fetal adverse effects.
The beneficial effects of antenatal steroids were similar in
studies conducted in the 1970s as in those conducted
tration [8], and there are no data to suggest a longer-term more recently implying that they remain beneficial in the
benefit (or harm) on infant and childhood outcomes [9]. presence of modern neonatal care [18]. Prenatal cortico-
Cervical cerclage may also reduce preterm birth in at-risk steroid therapy is recommended in all pregnancies with
pregnancies, but it is not clear whether it improves peri- threatened preterm labour before 34 weeks gestation
natal outcome [10]. Adequate spacing between pregnan- where active care of the newborn is anticipated. Although
cies may reduce the risk of recurrent preterm delivery; a there are limited RCT data in babies <26 weeks gestation
Caesarean delivery is likely to increase the risk of sponta- or very immature twins, observational studies support
neous preterm delivery in a subsequent pregnancy. the concept that antenatal corticosteroids also reduce
Interventions to prevent RDS and improve outcome mortality in these infants [19, 20]. In pregnancies be-
can also begin before birth even if delivery cannot be pre- tween 34 and 36 weeks gestation, prenatal steroids will
vented. There is often warning of impending preterm de- also reduce the risk of short-term respiratory morbidity
livery, and interventions can be considered that might but not mortality, and there is a paucity of data on longer-
prolong gestation or reduce the risk of an adverse out- term follow-up [21]. When given before elective Caesar-
come by preparing the fetus, or enabling transfer to a ean section (CS) at 3739 weeks, they reduce the risk of
centre with more experience of dealing with problems of admission to the NICU, although the number needed to
prematurity. Cervical length measurement, in combina- treat is >20 [22]. Follow-up data on term babies exposed
tion with fetal fibronectin testing, can help to determine to antenatal steroids are limited.
which women are at low risk of delivery within 7 days, The optimal treatment to delivery interval is more
and perhaps allow a more judicious use of antenatal treat- than 24 h and less than 7 days after the start of steroid
ments [11]. Extremely preterm babies at risk of RDS treatment; beyond 14 days the benefits are diminished.
should be born in centres where appropriate skills are There is a continuing debate as to whether steroids should
available, as long-term health outcomes are better if they be repeated 1 or 2 weeks after the first course for women
receive their initial neonatal care in tertiary units [12]. In with threatened preterm labour. Such repeat courses do
cases of prenatal prelabour rupture of membranes, anti- not reduce the risk of neonatal death, but reduce RDS and
biotics can delay preterm delivery and reduce neonatal other short-term health problems, although birth weight
morbidity including the need for surfactant, although co- is reduced and long-term beneficial effects are lacking
amoxiclav should be avoided because of an association [23]. The WHO recommends that a single repeat course
with an increased risk of necrotizing enterocolitis (NEC) of steroids may be considered if preterm birth does not
[13]. Magnesium sulphate given to women with immi- occur within 7 days after the initial course and subsequent
nent preterm delivery marginally reduces the incidence assessment demonstrates that there is a high risk of pre-
of cerebral palsy [14], although a more recent longer- term birth in the next 7 days [24]. It is unlikely that repeat
term follow-up of an Australian cohort showed no differ- courses given after 32 weeks gestation improve outcome,
ences by school age [15]. Tocolytic drugs can be used in and recent long-term follow-up studies show no benefit
the short term to delay birth and allow safe transfer to a by school age in terms of reduction in death or disability
perinatal centre or to enable prenatal corticosteroids time if repeat courses are used [25].
to take effect. However, no beneficial effects of tocolytic A recent RCT from low- to medium-income countries
drugs have been shown in randomized controlled trials showed a higher neonatal mortality and maternal infec-
158.232.240.113 - 9/26/2016 8:31:19 PM

European Consensus Guidelines on the Neonatology 2017;111:107125 109


Management of RDS 2016 Update DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
tion rate in women given prenatal steroids [26]. The ma- highlighting recent evidence-based approaches to assess-
jority of babies were >2 kg at birth, and these data empha- ing and supporting babies during the immediate postna-
size the importance of adequate dating of duration of tal period, including newborn life support where required
pregnancy, assessment of risk of preterm birth and avail- [28]. Resuscitation training courses tend to focus on ba-
ability of neonatal facilities before considering antenatal bies with terminal apnoea secondary to prolonged hypox-
steroids. Steroids are potent drugs with many potential ia with appropriate emphasis of achieving lung inflation
side effects. When given appropriately they improve out- by observing adequate chest lift, and judging a baby to be
come. If not, then side effects, such as impaired fetal and well when they are pink. When dealing with preterm ba-
placental growth, apoptosis in the brain and increased in- bies with RDS, we must train ourselves to think differ-
fection risks, may prevail. Use of steroids should be re- ently, allowing the infant to pass gradually through tran-
duced by adequate preterm birth risk assessment and by sition if possible whilst exposing them to a minimum
avoidance of early elective CS. In some cases when an number of interventions that may cause harm [29].
early CS is needed, establishment of fetal lung maturity Timing of clamping of the umbilical cord is important.
may be better than giving steroids to all women [27]. In Traditionally the cords of preterm infants were clamped
addition, there is no evidence that delivering preterm in- and cut immediately after birth to enable the paediatri-
fants by CS rather than allowing vaginal delivery im- cians to begin resuscitation as quickly as possible under a
proves outcome. radiant heater. Studies in cannulated fetal lambs showed
that cord clamping before lung aeration has occurred re-
Recommendations sults in acute transient reduction in left ventricular out-
1 Mothers at high risk of preterm birth <2830 weeks gesta-
tion should be transferred to perinatal centres with experi- put. Delaying clamping until the lungs are aerated and left
ence in the management of RDS (C1). atrial blood flow is established results in smoother transi-
2 Clinicians should offer a single course of prenatal corticoste- tion with no fluctuation in blood pressure [30]. Random-
roids to all women at risk of preterm delivery, from when ized trials show that promoting placentofetal transfusion
pregnancy is considered potentially viable up to 34 complet- results in a higher haematocrit, transiently higher blood
ed weeks gestation (A1).
3 A single repeat course of antenatal steroids may be appropri- pressure with less need for inotropic support and fewer
ate if the first course was administered more than 12 weeks intraventricular haemorrhages [31]. Umbilical cord milk-
previously and the duration of pregnancy is <3234 weeks ing in preterm babies may be an alternative to delayed
gestation when another obstetric indication arises (A2). cord clamping, particularly at CS or in emergency situa-
4 Antenatal steroids can also be considered for CS not in la- tions but concerns about safety remain, and there is a
bour up to 39 weeks (B2). However, there should be a clear
medical reason to do an early CS, and elective CS should not paucity of long-term follow-up data for either method
be performed <39 weeks gestation. [32, 33]. Our previous strong recommendation in sup-
5 In late preterm pregnancy at risk of early birth, a course of port of delaying cord clamping has been criticized, as ev-
antenatal steroids may also be considered provided there is idence on which it was based had relatively few extreme-
no evidence of chorio-amnionitis (C2). ly preterm babies and a paucity of longer-term follow-up
6 In women with symptoms of preterm labour, cervical length
and fibronectin measurements should be considered to pre- data [34]. A recent trial of 208 singleton fetuses <32 weeks
vent unnecessary hospitalization and use of tocolytic drugs gestation showed no difference in hospital outcomes, but
and/or antenatal steroids (B2). improved neurodevelopmental outcome at 18 months
7 Clinicians should consider short-term use of tocolytic drugs [35]. The Australian Placental Transfusion Study will
in very preterm pregnancies to allow completion of a course compare outcomes in 1,600 babies <30 weeks gestation
of prenatal corticosteroids and/or in utero transfer to a peri-
natal centre (B1). randomized to immediate or cord clamping delayed for
60 s and will hopefully provide a more definitive answer
[36]. After birth the baby should be placed in a clear poly-
ethylene bag and under a radiant warmer to maintain
Delivery Room Stabilization body temperature (see later).
Stabilization of preterm babies with RDS may require
Babies with RDS have difficulty maintaining alveolar inflation of the lung with blended air/oxygen, and how
aeration after birth, although most try to breathe for this should be done has been studied in some detail. Air
themselves, and therefore any support of transition is is better than oxygen for resuscitation of term babies in
stabilization rather than resuscitation. Updated Euro- terms of reduced mortality, and 100% oxygen is also
pean Resuscitation Guidelines were published in 2015, probably harmful to preterm babies, causing increased
158.232.240.113 - 9/26/2016 8:31:19 PM

110 Neonatology 2017;111:107125 Sweet etal.


DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
oxidative stress [37]. Protocols to achieve normal transi- 4 Intubation should be reserved for babies who have not re-
tional saturations measured by pulse oximetry at the right sponded to positive pressure ventilation via face mask (A1).
Babies who require intubation for stabilization should be giv-
wrist usually result in extremely low-birth-weight infants en surfactant (B1).
requiring around 3040% oxygen by about 10 min of age 5 Plastic bags or occlusive wrapping under radiant warmers
[38, 39]. Starting low and working up is better than start- should be used during stabilization in the delivery suite for
ing high and working down in terms of reducing oxida- babies <28 weeks gestation to reduce the risk of hypothermia
tive stress, although starting with 21% may be too low for (A1).
the most immature babies who may need at least 30%
oxygen, and further studies are under way to resolve this
issue [40]. Measuring heart rate by auscultation or cord Surfactant Therapy
palpation may not be accurate, and although ECG in the
delivery room offers a more rapid practical alternative to Surfactant therapy plays an important role in the man-
pulse oximetry for measuring heart rate, it is not widely agement of babies with RDS. By 2013 it was accepted that
available and may not offer any meaningful advantage in surfactant prophylaxis, in the current era of prenatal ste-
terms of improving outcome. The need to provide effec- roid use, was no longer indicated for babies receiving sta-
tive measurable CPAP from birth makes the T piece de- bilization using non-invasive respiratory support, and a
vice a better choice than a self-inflating anaesthetic bag strategy of initiation of CPAP from birth with early selec-
[41]. Routine suctioning is not needed before CPAP is tive surfactant administration for babies showing signs of
applied [42]. CPAP during stabilization can be delivered RDS was recommended, with the caveat that if the baby
either by face mask or a short nasal prong [43]. For spon- needed intubation for stabilization, surfactant should be
taneously breathing preterm babies, provision of CPAP given [4, 46]. The overall aim was to avoid mechanical
alone is optimal, and routine use of positive pressure ventilation (MV) where possible, or to reduce its dura-
breaths should be discouraged because of the risk of lung tion, whilst administering surfactant as early as possible
injury [44]. Gentle positive pressure ventilation should be in the course of RDS if it was deemed necessary. To this
provided for babies who remain apnoeic or bradycardic, end the INSURE (intubate-surfactant-extubate to CPAP)
and there is no apparent advantage of a sustained infla- technique was recommended with suggested protocols
tion over intermittent positive pressure breaths [45]. The for surfactant administration when babies showed signs
Sustained Aeration of Infants Lungs trial has been of RDS and needed more than 30% inspired oxygen to
launched and will hopefully more fully resolve this issue. maintain saturations in the normal range. Since the 2013
Only a minority of babies should require intubation for guideline update there have been further studies aimed at
stabilization. If intubation is required, the correct place- optimizing surfactant use, by avoiding potential exposure
ment of the endotracheal tube can be quickly verified to lung injury using less invasive methods of administra-
clinically by auscultation and using a colorimetric CO2 tion, and avoiding positive pressure ventilation through
detection device before administering surfactant which an endotracheal tube.
can be done prior to radiographic confirmation of RDS
in most circumstances. Surfactant Administration Methods
Surfactant administration is a skill that requires an ex-
Recommendations perienced clinical team comfortable with neonatal intu-
1 If possible delay clamping the umbilical cord for at least 60 s bation and MV if needed. Until recently the vast majority
to promote placentofetal transfusion (B1). Cord milking is a of clinical trials of surfactant used bolus administration
reasonable alternative if delayed cord clamping is not possi- via an endotracheal tube with a short period of manual
ble (B2). ventilation or MV to distribute the drug followed either
2 Oxygen for resuscitation should be controlled using a blend-
er. An initial concentration of 30% oxygen is appropriate for by continued MV or immediate (or early) extubation to
babies <28 weeks gestation, and 2130% for those of 2831 CPAP when spontaneous breathing had resumed if the
weeks, and adjustments up or down should be guided by INSURE method was used. INSURE was recommended
pulse oximetry from birth (B2). in the 2013 guideline on the basis that it reduced lung in-
3 In spontaneously breathing babies, stabilize with CPAP of at jury [47]; however, in the original studies sedation for in-
least 6 cm H2O via mask or nasal prongs (A1). Gentle posi-
tive pressure lung inflations using about 2025 cm H2O peak tubation was considered optional, and this was an area for
inspiratory pressure should be used for persistently apnoeic debate. Since then there have been studies to determine if
or bradycardic infants (B1). surfactant administration without endotracheal intuba-
158.232.240.113 - 9/26/2016 8:31:19 PM

European Consensus Guidelines on the Neonatology 2017;111:107125 111


Management of RDS 2016 Update DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
Table 2. Surfactant preparations (animal-derived) licensed in Europe in 2016

Generic name Trade name Source Manufacturer Dose (volume)

Beractant Survanta Bovine Ross Laboratories (USA) 100 mg/kg/dose (4 ml/kg)


Bovactant Alveofact Bovine Lyomark (Germany) 50 mg/kg/dose (1.2 ml/kg)
Poractant alfa Curosurf Porcine Chiesi Farmaceutici (Italy) 100 200 mg/kg/dose (1.25 2.5 ml/kg)

tion results in improved outcomes, on the basis that tants are better than older synthetic (protein-free) prepa-
avoidance of any positive pressure ventilation may be rations, containing only phospholipids, at reducing pul-
beneficial. Two similar methods of administering surfac- monary air leaks and mortality [55]. Lucinactant is a syn-
tant via a fine catheter without traditional intubation thetic surfactant that contains sinapultide, a protein
have been studied. The first, developed in Germany and analogue mimicking surfactant protein-B (SP-B) activity.
now used widely in parts of Europe, uses a fine flexible It works better than the protein-free synthetic surfac-
catheter positioned in the trachea whilst the baby is kept tants, but is not yet proven to be better than animal-de-
on CPAP, using laryngoscopy and Magills forceps [48], rived surfactants and is not available in Europe [56]. Syn-
also known as LISA (less invasive surfactant administra- thetic surfactants containing both SP-B and SP-C ana-
tion). The second, developed in Australia, uses a more logues are also currently under evaluation in clinical trials
rigid thin vascular catheter that is stiff enough to be posi- [57]. Comparisons among animal-derived surfactants
tioned in the trachea under direct laryngoscopy without have also shown differences in clinical effect. Overall
forceps whilst the baby is kept on CPAP [49], also known there is a survival advantage when a 200 mg/kg dose of
as MIST (minimally invasive surfactant treatment). With poractant alfa is compared with 100 mg/kg of beractant
both methods the aim is to maintain spontaneous breath- or 100 mg/kg poractant alfa to treat RDS but it is unclear
ing on CPAP whilst surfactant is gradually administered whether this is a dose effect or related to differences in the
over several minutes using a syringe without resorting to surfactant preparations [58].
routine bagging. Both of these methods have been com-
pared to traditional intubation for surfactant administra- When to Treat with Surfactant?
tion followed by MV. Large cohort studies from the Ger- Where possible babies at risk of RDS should be started
man neonatal network with experience of this method on CPAP from birth, and where possible maintained on
were encouraging, reporting reduced use of MV and less CPAP without resorting to intubation. If RDS develops,
BPD [50]. A randomized non-blinded clinical trial of ex- and surfactant is needed, the earlier in the course of the
tremely preterm infants between 23 and 27 weeks gesta- disease surfactant is given, the better the outcome, in
tion showed no significant increase in survival without terms of reducing air leaks [59], or maximizing the chance
BPD in those treated with LISA although these infants of successful avoidance of MV if the INSURE technique
required less ventilation, had fewer pneumothoraces and is used [60]. However, prophylactic INSURE does not
a reduction in severe intraventricular haemorrhage. confer any advantage over initiation of CPAP alone [61].
However, nearly 75% of the intervention group eventu- The previous guideline recommendation was that surfac-
ally needed MV, and the rate of desaturations was sig- tant should be administered when FiO2 >0.30 for very im-
nificantly higher in this group [51]. Although direct com- mature babies and >0.40 for more mature infants, based
parison with INSURE reported improved outcomes in on thresholds used in trials comparing earlier versus later
one study [52], reanalysis of the data could not reproduce surfactant from an era when CPAP was not in widespread
statistical significance when included in a meta-analysis, use. Recent observational studies have confirmed that
and therefore to date this is still uncertain [53]. Nebuliza- FiO2 >0.30 by 2 h of age on CPAP is predictive of CPAP
tion to deliver surfactant has not yet reached a stage where failure by 6 h of age, and that those who fail CPAP have a
it can be recommended for routine clinical use [54]. poorer outcome [62]. This strengthens the argument for
interventions that reduce CPAP failure, such as early sur-
Surfactant Preparations factant given by minimally invasive methods, to avoid
Surfactants currently available in Europe are shown in lung injury. Methods to measure the presence or absence
table2. Animal-derived (formerly called natural) surfac- of endogenous surfactant, such as lamellar body count in
158.232.240.113 - 9/26/2016 8:31:19 PM

112 Neonatology 2017;111:107125 Sweet etal.


DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
gastric aspirate, may help in deciding whom to treat [63]. in a prospective meta-analysis to give greater power to
However, methods that require laboratory expertise detect relatively small but important differences in out-
around the clock are unlikely to be widely adopted and a comes such as mortality [67]. Each trial was designed to
simple bedside test that can be used within the NICU is assess effects of saturation targeting in a lower range (85
needed. 89%) compared to a higher range (9195%) with blinding
There may be a need for further doses of surfactant. provided by oximeters which were offset to read higher
Randomized trials in the era before non-invasive ventila- or lower than the actual saturation within the target range.
tion was used widely showed that multiple doses reduced The SUPPORT trial from the USA, the first to be com-
the risk of air leaks [64] but this may not be true in the era pleted, showed the low saturation target group had a halv-
of early CPAP. Using a larger dose of 200 mg/kg porac- ing of ROP in survivors, but a worrying 4% increase in
tant alfa for the first dose will reduce the need for redosing mortality [68]. An interim meta-analysis combining these
[58]. Multiple INSURE has also been successfully em- data and all accrued at that time from the UK and Aus-
ployed and does not appear to worsen outcomes [65]. tralian BOOST-II trials was undertaken at the request of
Predicting who is likely to fail INSURE using clinical cri- the data safety-monitoring committee. This confirmed
teria and blood gases may help define a population that the excess mortality when targeting lower saturations,
would be reasonable to maintain on MV [66]. and further enrolment was stopped in the UK and Aus-
tralia/New Zealand [69]. In the meantime the Canadian
Recommendations COT trial was published reporting no significant differ-
1 Babies with RDS should be given a natural surfactant prepa-
ration (A1). ences in death or significant neurodisability, nor any sig-
2 A policy of early rescue surfactant should be standard (A1) nificant difference in ROP rates [70]. The outcomes from
but there are occasions when surfactant should be adminis- a combined analysis of the UK and Australia/New Zea-
tered in the delivery suite, such as those who require intuba- land BOOST-II trials have recently been published show-
tion for stabilization (B1). ing increased mortality in the low-saturation target group
3 Babies with RDS should be given rescue surfactant early in
the course of the disease. A suggested protocol would be to [71]. Meta-analysis of all available data in 2014 has fur-
treat babies 26 weeks gestation when FiO2 requirements ther added to uncertainty around ideal saturation targets.
>0.30 and babies >26 weeks when FiO2 requirements >0.40 Combining the data from 5 studies showed no difference
(B2). in death or disability at 24 months, no difference in mor-
4 Poractant alfa in an initial dose of 200 mg/kg is better than tality at 24 months but an increase in mortality before
100 mg/kg of poractant alfa or beractant for rescue therapy
(A1). hospital discharge in the restricted oxygen group [72].
5 INSURE should be considered for infants who are failing on Rates of ROP were similar in the two groups, although
CPAP (A2). there was an excess of NEC in the lower saturation group
6 LISA or MIST may be used as alternatives to INSURE for [72]. In 2016 the most up-to-date NeOProM meta-anal-
spontaneously breathing infants (B2). ysis confirmed that the lower target range (8589%) was
7 A second and sometimes a third dose of surfactant should be
administered if there is evidence of ongoing RDS such as per- associated with a significant increased risk of death but
sistent oxygen requirement and need for MV (A1). there was no difference between the 2 target ranges in
terms of disability at 1824 months [73]. Also, the lower
target range did not reduce BPD or severe visual impair-
ment but it did increase the risk of NEC requiring sur-
Oxygen Supplementation beyond Stabilization gery or causing death [73]. Current best evidence suggests
that it is reasonable to recommend aiming for saturations
For the last decade there has been an enormous effort between 90 and 94% rather than lower, although there is
to determine optimal saturation targets for preterm in- still a high level of uncertainty where ideal saturation tar-
fants, addressing the balance between avoiding negative gets lie.
effects of excess oxygen exposure such as retinopathy of The ability to maintain saturations within the pre-
prematurity (ROP) and potential negative effects of pro- defined target may also be important. There is a tension
longed low-grade hypoxia such as increased mortality, between narrower alarm limits, which increase alarm fre-
NEC or adverse neurodevelopmental outcome. The NeO- quency, nurse fatigue and potential wider fluctuations in
ProM collaboration was established to enable the results saturation and wider limits which may expose infants to
of 5 large-scale randomized clinical trials with similar potentially longer periods of time outside the desired
study design from different parts of the world to be used range [74]. Even in the best units many babies spend
158.232.240.113 - 9/26/2016 8:31:19 PM

European Consensus Guidelines on the Neonatology 2017;111:107125 113


Management of RDS 2016 Update DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
much time outside the target range, more usually hyper- single prongs although one small study suggested that na-
oxic than hypoxic [75, 76]. Secondary analysis of data sal masks may be the most effective interface for ensuring
from the COT trial oximeters showed an association of CPAP success [83]. All these interfaces have to be applied
prolonged episodes of hypoxaemia (saturation <80% for tightly to the face and carry a risk of facial distortion and
>1 min) with later death or adverse neurodevelopmental nasal trauma.
outcome [77]. Servo-controlled oxygen delivery algo- Bilevel CPAP is another variant of CPAP, or low-pres-
rithms show promise in terms of maintaining babies sure NIPPV, that uses small pressure differences between
within the target range for proportionately more time; inspiratory and expiratory phases. These are typically de-
however, to date no studies have been done to see if this livered through CPAP flow driver devices and generate
can improve outcome [78]. low peak inspiratory pressures of about 911 cm H2O
which can be synchronized using an abdominal pressure
Recommendations transducer. It is unclear whether these equate to changes
1 In preterm babies receiving oxygen, the saturation target
should be between 90 and 94% (B2). in tidal volume or simply an overall increase in the CPAP
2 To achieve this, suggested alarm limits should be 89 and 95% level. Although increasingly popular, there is not much
(D2). evidence that it confers any significant advantage over
CPAP [84, 85].
NIPPV is also used as first- or second-line respiratory
Non-Invasive Respiratory Support support in many units, with conventional ventilators
used to deliver peak inspiratory pressures similar to those
Non-invasive respiratory support is considered the on MV, with or without synchronization, but through na-
optimal method of providing assistance to preterm babies sal prongs [86]. NIPPV reduces extubation failure, but
with breathing problems and includes CPAP, various has not consistently been beneficial in reducing BPD [87].
types of ventilation provided through soft nasal prongs or Studies where NIPPV was most successful used synchro-
masks which are collectively called nasal intermittent nization of inspiratory pressure delivered through a sig-
positive pressure ventilation (NIPPV) and humidified nal from an abdominal Graseby capsule. These ventila-
oxygen delivered by high-flow nasal cannulae (HF). They tors are not widely available and delivering effective syn-
are used where possible as a substitute for MV as they are chronization using flow sensors is challenging due to
less injurious to the lung. Traditionally non-invasive large leaks during CPAP, and it is unclear whether non-
methods were used as a step-down from MV through an synchronized NIPPV is effective [86, 87]. The NIPPV tri-
endotracheal tube, and initially CPAP was the main al was a large international multicentre randomized trial
method employed, with early randomized trials showing powered to study the outcome of BPD in 1,009 babies
reduction in need for re-intubation if CPAP was used in- <1,000 g birth weight without specifying the mode of de-
stead of head box oxygen [79]. More recently it has been livering NIPPV, and it showed no difference between ba-
shown that initiation of CPAP from birth rather than rou- bies randomized to NIPPV compared with CPAP [88].
tine intubation for stabilization or prophylactic surfac- Planned secondary analysis of data from the NIPPV trial
tant treatment is better at preventing lung injury, al- also shows no difference in rates of BPD or death when
though it is important to try to determine when CPAP comparing those who received NIPPV compared to bi-
alone is not going to be effective [80]. CPAP devices pro- level CPAP [89]. Further work is needed to determine the
vide a flow of gas under controlled pressure delivered to best method of delivering NIPPV and the population
the nose via interfaces that are applied tightly to the face most likely to benefit.
to create a seal. Distending pressure has several theoreti- Since the 2013 guideline, the use of heated humidified
cal benefits including splinting the upper airway, main- HF as an alternative to CPAP has increased in popularity.
taining lung expansion and preventing end-expiratory A recent meta-analysis of 15 studies comparing HF with
alveolar collapse, thereby facilitating endogenous surfac- other modes of non-invasive respiratory support is reas-
tant release [81]. There seem to be no differences among suring, showing equivalent rates of treatment failure for
devices used to deliver CPAP pressure, although the in- babies coming off MV and similar rates of BPD, although
terface may be important [82]. In the delivery room a there is still a relative paucity of data for the extremely
short pharyngeal tube is a reasonable alternative to face preterm population, and wide confidence intervals and
mask CPAP to free up hands during early stabilization heterogeneity in relation to equivalence for treatment
[43]. In the NICU short binasal prongs are better than failure [90]. A potential mechanism of benefit may be
158.232.240.113 - 9/26/2016 8:31:19 PM

114 Neonatology 2017;111:107125 Sweet etal.


DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
carbon dioxide washout of the nasopharyngeal space; ance after lung fluid clearance or surfactant administra-
however, with higher flow rates there is also an element tion. Excessively high tidal volumes injure the lung and
of unquantified additional CPAP. Flow rates of 4.08.0 lead to hypocarbia which can subsequently cause brain
l/min are typically used, with weaning of flow rate deter- injury such as periventricular leukomalacia or intraven-
mined clinically by FiO2 levels remaining low and judge- tricular haemorrhage [94, 95]. In contrast, inadequately
ment of work of breathing. HF is also being studied as a low tidal volumes, which can occur after a decrease in
primary mode of respiratory support in the delivery room lung compliance, cause uneven distribution of tidal vol-
[91], and the results of large trials comparing HF with umes, increase work of breathing, agitation of the infant
CPAP are awaited [92]. and hypercarbia. VTV enables clinicians to ventilate with
less variable tidal volumes and real-time weaning of pres-
Recommendations sure as lung compliance improves. There are different
1 CPAP should be started from birth in all babies at risk of
RDS, such as those <30 weeks gestation who do not need ways of VTV regulation, such as inspiratory pressure,
intubation for stabilization (A1). flow or time adjustment, but better tidal volume stability
2 The system delivering CPAP is of little importance; however, is a final result of all these ventilator-driven algorithms.
the interface should be short binasal prongs or a mask, and a VTV compared to PLV may reduce BPD or death and
starting pressure of about 68 cm H2O should be applied intraventricular haemorrhage, and shorten duration of
(A2). CPAP pressure can then be individualized depending
on clinical condition, oxygenation and perfusion (D2). MV [96, 97]. Both an initial set tidal volume of about
3 CPAP with early rescue surfactant should be considered the 5 ml/kg in VTV and an estimated peak inspiratory pres-
optimal management for babies with RDS (A1). sure according to observation of chest movement in PLV
4 Synchronized NIPPV, if delivered through a ventilator rath- may need to be adjusted according to the babys own res-
er than a bilevel CPAP device, can reduce extubation failure, piratory efforts and gas exchange assessment. The re-
but may not confer long-term advantages such as reduction
in BPD (B2). quired tidal volume may need to increase with increasing
5 HF may be used as an alternative to CPAP for some babies postnatal age if the baby remains ventilated [98]. An
during the weaning phase (B2). open lung is achieved by setting adequate PEEP, and this
must be adjusted according to lung biophysical proper-
ties [99]. To determine optimum PEEP on conventional
Mechanical Ventilation Strategies ventilation, each incremental change of PEEP should be
evaluated by responses in FiO2 and CO2 levels. Lung com-
Despite best intentions to manage preterm babies on pliance is very dynamic during the management of RDS,
non-invasive respiratory support to protect their lungs, particularly following surfactant therapy, and for an indi-
about half of extremely preterm babies with RDS will fail vidual baby rapid ventilator responses may be needed.
and need support with MV through an endotracheal tube When high pressures are needed to achieve adequate
[93]. The aim of MV is to provide acceptable blood gas lung inflation, high-frequency oscillatory ventilation
exchange whilst minimizing the risk of lung injury, hypo- (HFOV) may be a reasonable alternative to MV. HFOV
carbia and circulatory disturbance. The principle of MV allows gas exchange with very low tidal volumes delivered
is to recruit atelectatic lung by inflation and optimize lung at very fast rates in lungs held open at optimal inflation
volume for an even distribution of tidal volumes at pres- by a continuous distending pressure. The optimum con-
sures set to prevent atelectasis and overinflation with tinuous distending pressure on HFOV is about 12 cm
minimal oxygen requirement. Overinflation increases the H2O above the closing pressure identified by deteriora-
risk of air leaks such as pneumothorax and pulmonary tion of oxygenation during stepwise reductions in airway
interstitial emphysema. However, ventilation at too low a pressure after full lung recruitment [100]. A recently up-
pressure risks areas of lung becoming repeatedly atelec- dated meta-analysis of RCTs comparing HFOV with con-
tatic during expiration, which can generate inflamma- ventional PLV shows a small inconsistent reduction in
tion. Modern neonatal ventilators offer various modes, BPD in the HFOV group; however, this benefit is coun-
with pressure-limited ventilation (PLV) and volume-tar- teracted by increased air leaks in those on HFOV [101].
geted ventilation (VTV) being the most common. Al- Although most trials reporting neurodevelopmental out-
though pressure-limited modes are relatively simple to come have not identified any differences, a recent long-
manage and allow ventilation even during the presence of term pulmonary follow-up of one RCT demonstrated
a large endotracheal tube leak, tidal volumes may increase better small airway function at 1114 years of age in in-
dangerously during rapid improvement of lung compli- fants originally managed with HFOV [102]. Whatever
158.232.240.113 - 9/26/2016 8:31:19 PM

European Consensus Guidelines on the Neonatology 2017;111:107125 115


Management of RDS 2016 Update DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
mode is accepted as standard within an individual unit, it outcomes such as BPD [108110]. Although this relation-
is important that all staff should be familiar with its use. ship cannot be assumed to be cause and effect, it seems
Overdistension should be considered if a baby is dete- reasonable in the absence of randomized trials and good
riorating on MV following surfactant administration, or evidence of safety to recommend caffeine routinely as
when an increase in mean airway pressure is followed by part of a strategy to minimize the need for MV. The stan-
increasing oxygen requirement. During ventilation hypo- dard dose of caffeine citrate is 20 mg/kg loading and 510
carbia and severe hypercarbia should be avoided because mg/kg daily maintenance. Some studies suggest that dou-
of their association with an increased risk of BPD, peri- bling these doses may further reduce the risk of extuba-
ventricular leukomalacia and intraventricular haemor- tion failure, although tachycardia is more frequent [111,
rhage, and methods of continuous CO2 assessment can be 112].
helpful during initiation of ventilation. When satisfactory
gas exchange is achieved and spontaneous breathing is Permissive Hypercarbia
present, weaning of ventilation should be started imme- In the 2013 guideline moderate hypercarbia was con-
diately. The VTV mode enables automatic weaning by a sidered tolerable during weaning from MV provided pH
decrease in peak inspiratory pressure in real time as com- was acceptable (above 7.22) in order to reduce time spent
pliance improves. Some babies will only require ventila- on MV [113]. More recently post hoc analysis of data
tion for a very short period of time. Babies with RDS im- from the SUPPORT trial shows an association between
prove rapidly following surfactant therapy and can be higher PaCO2 and risk of death, intraventricular haemor-
quickly weaned to low ventilator settings with a view to rhage, BPD and adverse neurodevelopmental outcome,
an early trial of extubation to CPAP. Early extubation of again highlighting the need for further evaluation of ide-
even the smallest babies is encouraged, provided it is al PaCO2 targets [95]. The PHELBI trial randomized ven-
judged clinically safe and they have acceptable blood gas- tilated preterm babies <29 weeks gestation and <1,000 g
es on low ventilator settings [103]. Extubation may be birth weight to two target PaCO2 levels for the first 14 days
successful from 78 cm H2O mean arterial pressure on of ventilation, the higher arm reaching about 10 kPa and
conventional modes and from 89 cm H2O continuous the lower about 8 kPa [114]. The study was stopped early
distending pressure on HFOV. Keeping stable very pre- and analysis performed on 359 of a planned 1,534 infants.
term babies on low-rate MV for longer periods does not There was no difference in the primary outcome of death
improve the chance of successful extubation [104]. Extu- or BPD, with trends to worse outcomes in the higher tar-
bating to a relatively higher level of CPAP pressure of 7 get group, including an increase in NEC in the smallest
9 cm H2O will improve the chance of successfully remain- babies and more death or BPD in infants with initial se-
ing off the ventilator [105]. vere lung disease in the higher target group. Tolerating
Several strategies have been used specifically to im- moderate hypercarbia to the level of the lower target
prove the success of non-invasive ventilation and shorten group of the PHELBI trial seems reasonable.
the duration of MV including caffeine therapy, permis-
sive hypercarbia and postnatal steroid treatment. Postnatal Steroids
One of the key objectives of RDS management is to
Caffeine Therapy improve survival whilst preventing BPD, and although
Since the 2010 guideline, caffeine therapy has been management of BPD is beyond the remit of this guideline,
recommended as an essential part of newborn respiratory it is worth considering strategies that can reduce lung in-
care [3]. The Caffeine for Apnea of Prematurity (CAP) flammation during the acute stage of RDS and potential-
study showed that caffeine facilitated earlier extubation ly limit the time on MV. Postnatal dexamethasone reduc-
with a significant reduction in BPD, and follow-up at 18 es BPD but its use declined dramatically when it was as-
months showed a reduction in neurodisability [106, 107]. sociated with an increased risk of cerebral palsy [115].
Caffeine was strongly recommended for babies with RDS However, BPD is also associated with adverse neurologi-
coming off ventilation and also for babies on non-inva- cal outcome and the higher the risk of BPD, the more po-
sive support to reduce risk of apnoea, although this was tential benefit there will be from a course of postnatal ste-
less evidence based, as the CAP trial had relatively few roids [116]. Low-dose dexamethasone (<0.2 mg/kg day)
infants who were treated prophylactically. Recently there is currently recommended for babies who remain ventila-
have been several large cohort studies supporting the use tor dependent after 12 weeks [117], and work is ongoing
of earlier rather than later caffeine in terms of improving to determine if even lower doses are effective [118]. Low-
158.232.240.113 - 9/26/2016 8:31:19 PM

116 Neonatology 2017;111:107125 Sweet etal.


DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
dose hydrocortisone also has the potential to reduce BPD, anticipated there will be need for regular blood gas analy-
but again long-term follow-up data are needed before this ses. Transcutaneous oxygen and CO2 monitoring has also
therapy can be routinely recommended [119]. Inhaled been used to access continuous information for trending
budesonide seems an obvious logical alternative to sys- but can cause skin injury [123]. Methods of monitoring
temic steroids and recently a large RCT confirmed that cerebral oxygenation are also available with potential to
prophylactic inhaled budesonide reduces both persistent guide clinicians about optimal cerebral blood flow but no
ductus arteriosus (PDA) and BPD [120]. There was a clear clinical benefit has yet been identified [124]. Access
trend toward increased mortality with treatment, and to laboratory support is necessary to enable close moni-
long-term developmental follow-up is not yet available. toring of serum electrolytes and haematological values
The addition of budesonide to natural surfactant prepa- ideally using microsampling techniques. Blood pressure
rations may also decrease lung inflammation in ventilat- should be recorded via indwelling arterial lines or inter-
ed preterm babies and reduce the risk of BPD, although mittently using approved oscillometric devices. Around-
this will need further verification by randomized multi- the-clock access to radiology services and portable ultra-
centre trials [121]. sound is also essential as these are often used to confirm
RDS diagnosis, exclude air leaks and confirm correct
Recommendations placement of endotracheal tubes and central lines.
1 After stabilization, MV should be used in babies with RDS
when other methods of respiratory support have failed (A1).
Duration of MV should be minimized (B2). Temperature Control
2 Targeted tidal volume ventilation should be employed as this Maintaining normal body temperature during stabili-
shortens the duration of ventilation and reduces BPD and zation and after admission is important for babies with
intraventricular haemorrhage (A1). RDS. The most recent International Liaison Committee
3 Avoid hypocarbia (A1) as well as severe hypercarbia (C2) as
these are associated with an increased risk of brain injury. on Resuscitation guidelines for resuscitation recom-
When weaning from MV, it is reasonable to tolerate a modest mend maintaining body temperature between 36.5 and
degree of hypercarbia, provided the pH remains above 7.22 37.5 C, and suggest that to do this effectively in preterm

(B2). babies requires environmental temperature in the deliv-


4 Caffeine should be used to facilitate weaning from MV (A1). ery room to be above 25 C [28]. Initial stabilization

Early caffeine should be considered for all babies at high risk


of needing MV, such as those <1,250 g birth weight, who are should be performed with the baby wrapped in a poly-
managed on non-invasive respiratory support (C1). ethylene bag under a radiant warmer [125]. Addition of
5 A short tapering course of low-dose dexamethasone should an exothermic mattress may increase the risk of over-
be considered to facilitate extubation in babies who remain heating [126]. Warming and humidification of gases
on MV after 12 weeks (A2). used for stabilization may also improve temperature
6 Inhaled steroids cannot be recommended for routine use to
reduce BPD until further safety data become available. [127]. Following stabilization infants should be nursed in
incubators with high relative humidity to reduce insen-
sible water losses. Servo-controlled incubators with skin
temperature set at 36.5 C decrease neonatal mortality

Monitoring and Supportive Care [128]. For the smallest babies humidity of 6080% should
be used initially and reduced as skin integrity improves,
To achieve best outcomes for preterm babies with RDS as maintaining high humidity may promote bacterial or
they should have optimal supportive care, with monitor- fungal growth. WHO guidelines promote the use of kan-
ing physiological variables and appropriate responses. garoo mother care in stable low-birth-weight babies as a
The ability to maintain body temperature from birth is means of maintaining temperature and reducing mortal-
extremely important. Pulse oximetry and possibly ECG ity in lower income settings, and increasingly kangaroo
monitoring from birth provide rapid information of re- mother care is being used to maintain temperature to
sponses to stabilization [122]. In the NICU there should maximize maternal-infant bonding, even in babies on
be access to continuous pulse oximetry, ECG monitoring MV [129, 130].
as well as monitoring PaCO2 levels. Detection of exhaled
CO2 can ensure correct placement of endotracheal tubes, Recommendation
and continuous measurement of end-tidal CO2 also gives 1 Maintain core temperature between 36.5 and 37.5 C at all

useful information showing trends in gas exchange. Um- times (C1).


bilical or radial arterial cannulation is indicated if it is
158.232.240.113 - 9/26/2016 8:31:19 PM

European Consensus Guidelines on the Neonatology 2017;111:107125 117


Management of RDS 2016 Update DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
Early Fluids and Nutritional Support laxis has the potential to do more harm than good [141
During transition after birth, fluid management is 143]. Guidelines usually offer advice on when to screen
challenging. The smallest infants have very high initial for sepsis based on additional risk factors such as mater-
transcutaneous losses of water, and water and sodium nal chorio-amnionitis or early signs of septicaemia in the
move from the interstitial to the intravascular compart- hope that antibiotics are only prescribed for those at
ments. Typically fluids are initiated at about 7080 ml/kg/ greatest risk [144]. If screening for sepsis is necessary,
day and adjustments individualized according to fluid then antibiotics should be started empirically whilst wait-
balance, weight change and serum electrolyte levels. A ing for test results, such as negative blood cultures at 36
modest early postnatal weight loss is normal. Regimens 48 h and negative serial C-reactive protein measurements,
with more restricted compared with more liberal fluids before stopping them. At present it is reasonable not to
have better outcomes, with reductions in PDA, NEC and use routine antibiotics in preterm babies with RDS at low
BPD [131]. Delaying introduction of sodium supplemen- risk such as planned delivery by elective CS. For those
tation until beyond the third day or 5% weight loss will who have been started empirically on antibiotics, the
also improve outcome [132]. Nutrition should be started shortest possible course should be used.
immediately after stabilization. Enteral feeding volumes
will initially be limited, so parenteral nutrition should be Recommendation
1 Antibiotics are often started in babies with RDS until sepsis
used. Early initiation of amino acids results in positive has been ruled out, but policies should be in place to narrow
nitrogen balance [133], reduced time to regain birth the spectrum and minimize unnecessary exposure. A com-
weight and enhanced weight gain at discharge [134]. mon regimen includes penicillin or ampicillin in combina-
Higher phosphorus and potassium intakes may be need- tion with an aminoglycoside (D2). Antibiotics should be
ed in cases of enhanced amino acid supply [135]. Paren- stopped as soon as possible once sepsis has been excluded
(C1).
teral lipids should also be started from day 1 [136]. For
stable infants a small amount (0.51 ml/kg/h) of breast
milk can be started early to promote maturation of the
intestinal tract [137]. There is no evidence of increased Managing Blood Pressure and Perfusion
NEC with early initiation of feeds or advancing feeds
more rapidly up to 30 ml/kg/day in stable very low-birth- Hypotension and low systemic blood flow are associ-
weight babies [138, 139]. Mothers milk is the preferred ated with adverse long-term outcome, although the two
option for initiation of feeding; however, if not available are not always closely correlated [145]. Blood pressure is
then pasteurized donor breast milk is better than formu- lower with decreasing gestation and increases gradually
la as it reduces the risk of NEC [140]. over the first 24 h of life, but varies widely at each gesta-
tional age [146]. Defining hypotension as a mean arterial
Recommendations pressure less than gestational age in weeks is widely ac-
1 Most babies should be started on intravenous fluids of 7080
ml/kg/day while being kept in a humidified incubator al- cepted; however, many babies with RDS will breach this
though some very immature babies may need more (B2). threshold and there is no evidence that treating numer-
Fluids must be tailored individually according to serum so- ically defined hypotension will influence outcome [147].
dium levels and weight loss (D1). Functional echocardiography can be used to assess car-
2 Sodium intake should be restricted over the first few days of diac output and look for evidence of poor systemic blood
life and initiated after onset of diuresis with careful monitor-
ing of fluid balance and electrolyte levels (B1). flow to help decide whether treatment is needed, al-
3 Parenteral nutrition should be started from birth. Protein though this skill is not available in many units [148]. Hy-
can be started at 22.5 g/kg/day from day 1 (B2). Lipids potension during RDS may be related to hypovolaemia,
should also be started from day 1 and quickly built up to 3.0 large left-to-right ductus or atrial shunts, or to myocar-
g/kg/day as tolerated (C2). dial dysfunction, and confirmation of low systemic blood
4 Enteral feeding with mothers milk should be started from
the first day if the baby is haemodynamically stable (B1). flow and its cause may help to guide appropriate treat-
ment. Hypovolaemia is probably overdiagnosed and can
be minimized by delaying cord clamping. Dopamine is
Antibiotics more effective than dobutamine at increasing blood
It had been considered good practice to screen babies pressure and can improve cerebral blood flow in hypo-
who present with early respiratory distress for infection; tensive infants [149], although dobutamine may be a
however, it is now known that routine antibiotic prophy- more rational choice during the transitional period due
158.232.240.113 - 9/26/2016 8:31:19 PM

118 Neonatology 2017;111:107125 Sweet etal.


DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
Table 3. Summary of recommendations

Prenatal care Preterm babies at risk of RDS should be born in centres where appropriate care, including MV, is available
Judicious antenatal assessment should include risk of preterm delivery and need for maternal corticosteroids if
the risk is moderate or high. Tocolytics can be used to allow time for steroids to take effect or for safe transfer
where appropriate
Delivery room Aim to delay cord clamping at birth by at least 1 min, or cut the cord long and milk towards the baby after birth
stabilization Stabilize the baby in a plastic bag under a radiant warmer to prevent heat loss
Gently support breathing using CPAP if possible, and, if inflations are needed, avoid excessive tidal volumes.
ECG and pulse oximetry can help guide heart rate response to stabilization. Start with about 21 30% oxygen and
titrate up or down as needed
Intubation at birth should be considered only for those not responding to the above, although early intubation
and surfactant may be required for babies who demonstrate early signs of severe RDS such as chest retractions
and high oxygen requirements
Respiratory An animal-derived (natural) surfactant should be used and given as early as possible in the course of RDS. For
support and very immature infants, a treatment threshold of FiO2 0.30 0.40 on CPAP seems reasonable. Repeat doses of
surfactant surfactant may be required if there is ongoing evidence of RDS
Babies can often be extubated to CPAP or NIPPV immediately following surfactant, and judgement needs to be
made if an individual baby will tolerate this. Consider minimally invasive surfactant (LISA or MIST) as an
alternative to INSURE if your unit has appropriate expertise
For those who require MV, aim to ventilate for as short a time as possible, avoiding hyperoxia, hypocarbia and
volutrauma. This may be best achieved with VTV and saturation alarm limits set at 89 and 95%
Caffeine therapy should be used routinely to minimize the need for ventilation. Babies should be maintained on
non-invasive respiratory support in preference to MV if possible. Beyond 1 2 weeks, steroids should be
considered to facilitate extubation if the baby remains ventilated
In preterm babies receiving oxygen, the saturation target should be between 90 and 94%. To achieve this,
suggested alarm limits should be 89 and 95%
Supportive Maintain body temperature at 36.5 37.5 C at all times
care Start parenteral nutrition immediately with amino acids and lipids with initial fluid volumes about 70 80 ml/kg/
day for most babies and restrict sodium during the early transitional period
Enteral feeding with mothers milk should also be started on day 1 if the baby is stable
Antibiotics should be used judiciously and stopped early when sepsis is ruled out
Blood pressure should be monitored regularly, aiming to maintain normal tissue perfusion, if necessary using
inotropes. Hb should be maintained at acceptable levels
Protocols should be in place for monitoring pain and discomfort and consideration given for non-
pharmacological methods of minimizing procedural pain and judicious use of opiates for more invasive
procedures

to its ability to increase contractility and decrease after- these slightly more restrictive values [154]. However,
load [150]. Studies assessing different thresholds for in- long-term follow-up has shown some better cognitive
tervention with dopamine to determine if inotrope ther- outcomes in those with more liberal Hb thresholds [155],
apy influences long-term outcome are ongoing [151]. and further trials are ongoing to resolve this issue [156].
Epinephrine and hydrocortisone can be used in refrac- PDA may provide clinical problems for very preterm
tory hypotension when dopamine and dobutamine have babies with RDS in terms of low blood pressure, poor tis-
failed although there is little new evidence on safety or sue perfusion, pulmonary oedema and difficulty weaning
efficacy [152, 153]. from MV. As all infants start life with an open ductus ar-
Maintaining a reasonable haemoglobin (Hb) concen- teriosus, it is difficult to make recommendations for when
tration is also important. Randomized trials comparing to treat it. Surgical ligation of PDA is associated with
targeting more restrictive versus more liberal Hb concen- worse long-term neurodevelopmental outcome, and al-
trations (about 12 g/dl lower) result in less need for though it is not clear whether this is due to the PDA or its
blood transfusion without affecting hospital outcomes, treatment, surgery should only be considered after medi-
and suggested Hb thresholds for transfusion are based on cal therapy has failed [157]. Permissive tolerance of PDA
158.232.240.113 - 9/26/2016 8:31:19 PM

European Consensus Guidelines on the Neonatology 2017;111:107125 119


Management of RDS 2016 Update DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
is an acceptable strategy provided the infant is thriving, Miscellaneous
tolerating feeds and on minimal respiratory support Since the 2010 guidelines we have included a brief sec-
[158]. Cyclo-oxygenase inhibitors such as indomethacin tion on aspects of RDS management that arise infrequent-
or ibuprofen promote ductal closure, although ibuprofen ly. Each year new genetic mutations affecting surfactant
has fewer side effects [159]. More recently paracetamol systems are reported; these are usually fatal, and congeni-
has been shown to promote ductal closure although more tal SP-B and ABCA3 deficiency are beyond the scope of
trials with long-term follow-up are needed before it can this guideline. Surfactant therapy may also be useful in
be routinely recommended [160]. Early echocardiogra- situations where secondary surfactant inactivation occurs
phy-guided therapy of large PDAs is being studied as a such as meconium aspiration, congenital pneumonia and
means of improving outcomes whilst minimizing expo- pulmonary haemorrhage. There are few clinical trials sup-
sure to treatment [161, 162]. porting surfactant use in pneumonia [168], although in a
recent observational study babies with RDS complicated
Recommendations by pneumonia seemed to require more surfactant [169].
1 Treatment of hypotension is recommended when it is con-
firmed by evidence of poor tissue perfusion such as oliguria, Surfactant therapy improves oxygenation in babies with
acidosis and poor capillary return rather than purely on nu- pulmonary haemorrhage, and although there are no RCTs
merical values (C2). looking at outcomes compared with no treatment [170], a
2 Hb concentration should be maintained within normal lim- recent small trial comparing two different natural surfac-
its. A suggested Hb threshold for babies on respiratory sup- tant preparations in pulmonary haemorrhage showed
port is 11.5 g/dl (haematocrit 35%) in week 1, 10 g/dl (hae-
matocrit 30%) in week 2 and 8.5 g/dl (haematocrit 25%) be- more rapid improvement in oxygenation with poractant
yond 2 weeks of age (C2). alfa compared with beractant, but with no differences in
3 If a decision is made to attempt therapeutic closure of the other outcomes [171]. There are no data to support rou-
PDA, then indomethacin or ibuprofen have been shown to tine or rescue use of inhaled nitric oxide (iNO) in preterm
be equally efficacious: ibuprofen should be used as there is babies [172]. Despite this, iNO continues to be used in
less transient renal failure or NEC (A2).
many units, particularly for ill babies with severe respira-
tory failure and poor oxygenation [173, 174]. There is an
Pain and Sedation argument for rationalizing the use of iNO for specific pop-
Newborn babies can experience pain, and during man- ulations of preterm infants, for example those with pre-
agement of RDS it is important to consider the comfort mature rupture of membranes or documented pulmonary
of the baby. Procedures such as venepuncture, intubation hypertension and conducting further clinical trials [175,
and MV all have potential to cause discomfort, and it is 176]. Until these are completed, iNO cannot be recom-
good practice to have mechanisms for evaluating pain us- mended for use in preterm babies.
ing validated scoring systems [163]. Many clinicians pre-
fer to use a combination of a short-acting opiate, muscle Recommendations
1 Surfactant can be used for RDS complicated by congenital
relaxant and atropine to maximize comfort and improve pneumonia (C1).
the chances of successful intubation [164]. However, 2 Surfactant therapy can be used to improve oxygenation fol-
there is a balance between ensuring comfort during laryn- lowing pulmonary haemorrhage (C1).
goscopy and not oversedating infants when trying to 3 The use of iNO in preterm babies should be limited to those
maintain them on non-invasive respiratory support in clinical trials or those with severe hypoxaemia secondary
to documented pulmonary hypertension (D2).
[165]. Once stable on ventilation there is usually no need
for routine sedation [166]. Sucrose analgesia and other
non-pharmacological methods may be employed to re-
duce procedural pain [167]. Summary of Recommendations

Recommendations A summary of all recommendations is given in table3.


1 The routine use of morphine nfusions in ventilated preterm
infants is not recommended (C2).
2 Opioids should be used selectively, when indicated by clini-
cal judgement and evaluation of pain indicators (D1).
158.232.240.113 - 9/26/2016 8:31:19 PM

120 Neonatology 2017;111:107125 Sweet etal.


DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
Acknowledgements Disclosure Statement

A European panel of experts was convened under the auspices Henry L. Halliday and Christian P. Speer are consultants to
of the European Association of Perinatal Medicine to update evi- Chiesi Farmaceutici, Parma, the manufacturer of a leading animal-
dence-based guidelines on the management of RDS. The guide- derived surfactant preparation used to treat RDS and a caffeine
lines were prepared using evidence-based methods as summarized product for treatment of apnoea of prematurity. Henry L. Halliday
in table1. We are grateful to Roger Soll and Eric Shinwell for their and Christian P. Speer are joint Chief Editors of Neonatology.
helpful comments on the final draft of these guidelines.

References
1 Stoll BJ, Hansen NI, Bell EF, Walsh MC, Car- cebo-controlled trial on the effect of vaginal Research Network: Association of antenatal
lo WA, Shankaran S, et al; Eunice Kennedy micronized progesterone. Ultrasound Obstet corticosteroids with mortality, morbidity,
Shriver National Institute of Child Health and Gynecol 2011;38:272280. and neurodevelopmental outcomes in ex-
Human Development Neonatal Research 10 Alfirevic Z, Stampalija T, Roberts D, Jorgensen tremely preterm multiple gestation infants.
Network: Trends in care practices, morbidity, AL: Cervical stitch (cerclage) for preventing JAMA Pediatr 2016;170:593601.
and mortality of extremely preterm neonates, preterm birth in singleton pregnancy. Coch- 21 Gyamfi-Bannerman C, Thom EA, Blackwell
19932012. JAMA 2015;314:10391051. rane Database Syst Rev 2012;4: CD008991. SC, Tita AT, Reddy UM, Saade GR, et al: An-
2 Sweet D, Bevilacqua G, Carnielli V, Greisen 11 Van Baaren GJ, Vis JY, Wilms FF, Oudijk tenatal betamethasone for women at risk for
G, Plavka R, Saugstad OD, Simeoni U, et al; MA, Kwee A, Porath MM, et al: Predictive late preterm delivery. N Engl J Med 2016;374:
Working Group on Prematurity of the World value of cervical length measurement and fi- 13111320.
Association of Perinatal Medicine, European bronectin testing in threatened preterm labor. 22 Sotiriadis A, Makrydimas G, Papatheodorou
Association of Perinatal Medicine: European Obstet Gynecol 2014;123:11851192. S, Ioannidis JP: Corticosteroids for prevent-
consensus guidelines on the management of 12 Rautava L, Eskelinen J, Hkkinen U, Lehtonen ing neonatal respiratory morbidity after elec-
neonatal respiratory distress syndrome. J L; PERFECT Preterm Infant Study Group: tive caesarean section at term. Cochrane Da-
Perinat Med 2007;35:175186. 5-year morbidity among very preterm infants tabase Syst Rev 2009;4:CD006614.
3 Sweet DG, Carnielli V, Greisen G, Hallman in relation to level of hospital care. JAMA Pe- 23 Crowther CA, McKinlay CJD, Middleton P,
M, Ozek E, Plavka R, Saugstad OD, et al; Eu- diatr 2013;167:4046. Harding JE: Repeat doses of prenatal corti-
ropean Association of Perinatal Medicine: 13 Kenyon S, Boulvain M, Neilson JP: Antibiot- costeroids for women at risk of preterm
European consensus guidelines on the man- ics for preterm rupture of membranes. Coch- birth for improving neonatal health out-
agement of neonatal respiratory distress syn- rane Database Syst Rev 2013;12:CD001058. comes. Cochrane Database Syst Rev 2015;
drome in preterm infants 2010 update. Neo- 14 Doyle LW, Crowther CA, Middleton P, Mar- 7:CD003935.
natology 2010;97:402417. ret S, Rouse D: Magnesium sulphate for wom- 24 World Health Organization:WHO Recom-
4 Sweet DG, Carnielli V, Greisen G, Hallman en at risk of preterm birth for neuroprotection mendations on Interventions to Improve Pre-
M, Ozek E, Plavka R, Saugstad OD, et al; Eu- of the fetus. Cochrane Database Syst Rev term Birth Outcomes. Geneva, WHO, 2015.
ropean Association of Perinatal Medicine: 2009;1:CD004661. 25 Asztalos EV, Murphy KE, Willan AR, Mat-
European consensus guidelines on the man- 15 Darlow B, Austin N, French N, Campbell C, thews SG, Ohlsson A, Saigal S, et al: Multiple
agement of neonatal respiratory distress syn- Carse E, Hayes M, et al: School-age outcomes courses of antenatal corticosteroids for pre-
drome in preterm infants 2013 update. Neo- of very preterm infants after antenatal treat- term birth study: outcomes in children at 5
natology 2013;103:353368. ment with magnesium sulfate vs placebo. years of age (MACS-5). JAMA Pediatr 2013;
5 https://nightingale.vtoxford.org/reports.asp. JAMA 2014;312:11051113. 167:11021110.
6 Guyatt GH, Oxman AD, Kunz R, Falck-Ytter 16 Haas DM, Caldwell DM, Kirkpatrick P, Mc- 26 Althabe F, Belizn JM, McClure EM, Heming-
Y, Vist GE, Liberati A, Schnemann, HJ; Intosh JJ, Welton NJ: Tocolytic therapy for way-Foday J, Berrueta M, Mazzoni A, et al: A
GRADE Working Group: Going from evi- preterm delivery: systematic review and net- population-based, multifaceted strategy to
dence to recommendations. BMJ 2008; 336: work meta-analysis. BMJ 2012;345:e6226. implement antenatal corticosteroid treat-
10491051. 17 Tocolysis for women in preterm labour. ment versus standard care for the reduction of
7 Dodd JM, Jones L, Flenady V, Cincotta R, Green-top guideline No 1b, February 2011. neonatal mortality due to preterm birth in
Crowther CA: Prenatal administration of Royal College of Obstetricians and Gynaeco- low-income and middle-income countries:
progesterone for preventing preterm birth in logists. https://www.rcog.org.uk/globalas- the ACT cluster-randomised trial. Lancet
women considered to be at risk of preterm sets/documents/guidelines/gtg_1b.pdf. 2015;385:629639.
birth. Cochrane Database Syst Rev 2013; 18 Roberts D, Dalziel S: Antenatal corticoste- 27 American College of Obstetricians and Gyne-
7:CD004947. roids for accelerating fetal lung maturation cologists: ACOG committee opinion No 559:
8 Norman JE, Marlow N, Messow CM, Shen- for women at risk of preterm birth. Cochrane Cesarean delivery on maternal request. Ob-
nan A, Bennett PR, Thornton S, et al; Database Syst Rev 2006;3:CD004454. stet Gynecol 2013;121:904907.
OPPTIMUM study group: Vaginal progester- 19 Manktelow BN, Lal MK, Field DJ, Sinha SK: 28 Wyllie J, Bruinenberg J, Roehr CC, Rdiger
one prophylaxis for preterm birth (the Antenatal corticosteroids and neonatal out- M, Trevisanuto D, Urlesberger B: European
OPPTIMUM study): multicentre, random- comes according to gestational age: a cohort Resuscitation Council guidelines for resusci-
ised, double-blind trial. Lancet 2016; 387: study. Arch Dis Child Fetal Neonatal Ed 2010; tation 2015. Section 7: resuscitation and sup-
21062116. 95:F95F98. port of transition of babies at birth. Resuscita-
9 Rode L, Klein K, Nicolaides KH, Krampl-Bet- 20 Boghossian NS, McDonald SA, Bell EF, Carlo tion 2015;95:249263.
telheim E, Tabor A; PREDICT Group: Pre- WA, Brumbaugh JE, Stoll BJ, et al; Eunice 29 Saugstad OD: Delivery room management of
vention of preterm delivery in twin gestations Kennedy Shriver National Institute of Child term and preterm newly born infants. Neona-
(PREDICT): a multicenter, randomized, pla- Health and Human Development Neonatal tology 2015;107:365371.
158.232.240.113 - 9/26/2016 8:31:19 PM

European Consensus Guidelines on the Neonatology 2017;111:107125 121


Management of RDS 2016 Update DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
30 Bhatt S, Alison BJ, Wallace EM, Crossley KJ, nasal prong or face mask for respiratory sup- rived surfactant extract for the prevention
Gill AW, Kluckow M, te Pas AB, et al: Delay- port for preterm newborns. Pediatrics 2013; and treatment of respiratory distress syn-
ing cord clamping until ventilation onset im- 132:e389e395. drome. Cochrane Database Syst Rev 2007;
proves cardiovascular function at birth in pre- 44 Jobe AH, Ikegami M: Mechanisms initiating 4:CD006069.
term lambs. J Physiol 2013;591:21132126. lung injury in the preterm. Early Hum Dev 57 Curstedt T, Halliday HL, Speer CP: A unique
31 Rabe H, Diaz-Rossello JL, Duley L, Dowswell 1998;53:8194. story in neonatal research: the development
T: Effect of timing of umbilical cord clamping 45 ODonnell CP, Bruschettini M, Davis PG, of a porcine surfactant. Neonatology 2015;
and other strategies to influence placental Morley CJ, Moja L, Calevo MG, Zappettini S: 107:321329.
transfusion at preterm birth on maternal and Sustained versus standard inflations during 58 Singh N, Halliday HL, Stevens TP, Suresh G,
infant outcomes. Cochrane Database Syst Rev neonatal resuscitation to prevent mortality Soll R, Rojas-Reyes MX: Comparison of ani-
2012;8:CD003248. and improve respiratory outcomes. Cochrane mal-derived surfactants for the prevention
32 Katheria AC, Truong G, Cousins L, Oshiro B, Database Syst Rev 2015;7:CD004953. and treatment of respiratory distress syn-
Finer NN: Umbilical cord milking versus de- 46 American Academy of Pediatrics Committee drome in preterm infants. Cochrane Database
layed cord clamping in preterm infants. Pedi- on Fetus and Newborn: Respiratory support Syst Rev 2015;12:CD010249.
atrics 2015;136:6169. in preterm infants at birth. Pediatrics 2014; 59 Bahadue FL, Soll R: Early versus delayed se-
33 Al-Wassia H, Shah PS: Efficacy and safety of 133:171174. lective surfactant treatment for neonatal re-
umbilical cord milking at birth: a systematic 47 Stevens TP, Harrington EW, Blennow M, Soll spiratory distress syndrome. Cochrane Data-
review and meta-analysis JAMA Pediatr 2015; RF: Early surfactant administration with brief base Syst Rev 2012;11:CD001456.
169:1825. ventilation vs selective surfactant and contin- 60 Verder H, Albertsen P, Ebbesen F, Greisen G,
34 Tarnow-Mordi WO, Duley L, Field D, Mar- ued mechanical ventilation for preterm in- Robertson B, Bertelsen A, et al: Nasal contin-
low N, Morris J, Newnham J, et al: Timing of fants with or at risk for respiratory distress uous positive airway pressure and early sur-
cord clamping in very preterm infants: more syndrome. Cochrane Database Syst Rev 2007; factant therapy for respiratory distress syn-
evidence is needed. Am J Obstet Gynecol 4:CD003063. drome in newborns of less than 30 weeks ges-
2014;211:118123. 48 Gpel W, Kribs A, Ziegler A, Laux R, Hoehn tation. Pediatrics 1999;103:E24.
35 Mercer JS, Erickson-Owens DA, Vohr BR, T, Wieg C, et al: Avoidance of mechanical 61 Isayama T, Chai-Adisaksopha C, McDonald
Tucker RJ, Parker AB, Oh W, Padbury JF: Ef- ventilation by surfactant treatment of sponta- SD: Noninvasive ventilation with vs without
fects of placental transfusion on neonatal and neously breathing preterm infants (AMV): an early surfactant to prevent chronic lung dis-
18 month outcomes in preterm infants: a ran- open-label, randomised, controlled trial. Lan- ease in preterm infants: a systematic review
domized controlled trial. J Pediatr 2016; 168: cet 2011;378:16271633. and meta-analysis. JAMA Pediatr 2015; 169:
5055. 49 Dargaville PA, Aiyappan A, de Paoli AG, 731739.
36 https://www.anzctr.org.au/Trial/Registra- Kuschel CA, Kamlin CO, Carlin JB, Davis PG: 62 Agertoft L, Djernes B, Nathan E, Reinholdt J,
tion/TrialReview.aspx?id = 335752. Minimally invasive surfactant therapy in pre- Dargaville PA, Aiyappan A, et al: Continuous
37 Saugstad OD, Aune D, Aguar M, Kapadia V, term infants on continuous positive airway positive airway pressure failure in preterm in-
Finer N, Vento M: Systematic review and me- pressure. Arch Dis Child Fetal Neonatal Ed fants: incidence, predictors and consequenc-
ta-analysis of optimal initial fraction of oxy- 2013;98:F122F126. es. Neonatology 2013;104:814.
gen levels in the delivery room at 32 weeks. 50 Gpel W, Kribs A, Hrtel C, Avenarius S, Teig 63 Verder H, Ebbesen F, Fenger-Grn J, Henrik-
Acta Paediatr 2014;103:44751. N, Groneck P, et al: Less invasive surfactant sen TB, Andreasson B, Bender L, et al: Early
38 Vento M, Cubells E, Escobar JJ, Escrig R, administration is associated with improved surfactant guided by lamellar body counts on
Aguar M, Brugada M, et al: Oxygen saturation pulmonary outcomes in spontaneously gastric aspirate in very preterm infants. Neo-
after birth in preterm infants treated with breathing preterm infants. Acta Paediatr natology 2013;104:116122.
continuous positive airway pressure and air: 2015;104:241246. 64 Soll R, Ozek E: Multiple versus single doses of
assessment of gender differences and com- 51 Kribs A, Roll C, Gpel W, Wieg C, Groneck exogenous surfactant for the prevention or
parison with a published nomogram. Arch P, Laux R, et al: Nonintubated surfactant ap- treatment of neonatal respiratory distress
Dis Child Fetal Neonatal Ed 2013; 98:F228 plication vs conventional therapy in extreme- syndrome. Cochrane Database Syst Rev 2009;
F232. ly preterm infants: a randomized clinical trial. 1:CD000141.
39 Kapadia VS, Chalak LF, Sparks JE, Allen JR, Sa- JAMA Pediatr 2015;169:723730. 65 Dani C, Corsini I, Poggi C: Risk factors for
vani RC, Wyckoff MH: Resuscitation of pre- 52 Kanmaz HG, Erdeve O, Canpolat FE, Mutlu intubation-surfactant-extubation (INSURE)
term neonates with limited versus high oxygen B, Dilmen U: Surfactant administration via failure and multiple INSURE strategy in pre-
strategy. Pediatrics 2013;132:e1488e1496. thin catheter during spontaneous breathing: term infants. Early Hum Dev 2012; 88(suppl
40 Oei JL, Vento M, Rabi Y, Wright I, Finer N, randomized controlled trial. Pediatrics 2013; 1):S3S4.
Rich W, et al: Higher or lower oxygen for de- 131:e502e509. 66 Brix N, Sellmer A, Jensen MS, Pedersen LV,
livery room resuscitation of preterm infants 53 More K, Sakhuja P, Shah PS: Minimally inva- Henriksen TB: Predictors for an unsuccessful
below 28 completed weeks gestation: a meta- sive surfactant administration in preterm in- INtubation-SURfactant-Extubation proce-
analysis. Arch Dis Child Fetal Neonatal Ed fants: a meta-narrative review. JAMA Pediatr dure: a cohort study. BMC Pediatr 2014; 14:
2016, Epub ahead of print. 2014;168:901908. 155.
41 Szyld E, Aguilar A, Musante GA, Vain N, Pru- 54 Minocchieri S, Knoch S, Schoel WM, Ochs M, 67 Askie LM, Brocklehurst P, Darlow BA, Finer
dent L, Fabres J, Carlo WA; Delivery Room Nelle M: Nebulizing poractant alfa versus N, Schmidt B, Tarnow-Mordi W; NeOProM
Ventilation Devices Trial Group: Compari- conventional instillation: ultrastructural ap- Collaborative Group: NeOProM: Neonatal
son of devices for newborn ventilation in the pearance and preservation of surface activity. Oxygenation Prospective Meta-analysis Col-
delivery room. J Pediatr 2014;165:234239. Pediatr Pulmonol 2014;49:348356. laboration study protocol. BMC Pediatr 2011;
42 Kelleher J, Bhat R, Salas AA, Addis D, Mills 55 Ardell S, Pfister RH, Soll R: Animal derived 11:6.
EC, Mallick H, et al: Oronasopharyngeal suc- surfactant extract versus protein free synthet- 68 SUPPORT Study Group of the Eunice Ken-
tion versus wiping of the mouth and nose at ic surfactant for the prevention and treatment nedy Shriver NICHD Neonatal Research Net-
birth: a randomised equivalency trial. Lancet of respiratory distress syndrome. Cochrane work; Carlo WA, Finer NN, Walsh MC, Rich
2013;382:326330. Database Syst Rev 2015;8:CD000144. W, Gantz MG, Laptook AR, et al: Target rang-
43 McCarthy LK, Twomey AR, Molloy EJ, Mur- 56 Pfister RH, Soll R, Wiswell TE: Protein con- es of oxygen saturation in extremely preterm
phy JF, ODonnell CP: A randomized trial of taining synthetic surfactant versus animal de- infants. N Engl J Med 2010;362:19591969.
158.232.240.113 - 9/26/2016 8:31:19 PM

122 Neonatology 2017;111:107125 Sweet etal.


DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
69 BOOST II United Kingdom Collaborative respiratory distress syndrome: continuous 94 Erickson SJ, Grauaug A, Gurrin L, Swamina-
Group; BOOST II Australia Collaborative positive airway pressure and nasal intermit- than M: Hypocarbia in the ventilated pre-
Group; BOOST II New Zealand Collaborative tent positive pressure ventilation. Semin Fetal term infant and its effect on intraventricular
Group; Stenson BJ, Tarnow-Mordi WO, Dar- Neonatal Med 2009;14:1420. haemorrhage and bronchopulmonary dys-
low BA, Simes J, Juszczak E, Askie L, et al: Ox- 82 De Paoli AG, Davis PG, Faber B, Morley CJ: plasia. J Paediatr Child Health 2002;38:560
ygen saturation and outcomes in preterm in- Devices and pressure sources for administra- 562.
fants. N Engl J Med 2013;368:20942104. tion of nasal continuous positive airway pres- 95 Ambalavanan N, Carlo WA, Wrage LA, Das
70 Schmidt B, Whyte RK, Asztalos EV, Mod- sure (NCPAP) in preterm neonates. Co- A, Laughon M, Cotten CM, et al; SUPPORT
demann D, Poets C, Rabi Y, et al; Canadian chrane Database Syst Rev 2002;3:CD002977. Study Group of the NICHD Neonatal Re-
Oxygen Trial (COT) Group: Effects of target- 83 Kieran EA, Twomey AR, Molloy EJ, Murphy search Network: PaCO2 in surfactant, posi-
ing higher vs lower arterial oxygen satura- JF, ODonnell CP: Randomized trial of prongs tive pressure, and oxygenation randomised
tions on death or disability in extremely pre- or mask for nasal continuous positive airway trial (SUPPORT). Arch Dis Child Fetal Neo-
term infants: a randomized clinical trial. pressure in preterm infants. Pediatrics 2012; natal Ed 2015;100:F145F149.
JAMA 2013;309:21112120. 130:e1170e1176. 96 Peng W, Zhu H, Shi H, Liu E: Volume-tar-
71 BOOST-II Australia and United Kingdom 84 Lampland AL, Plumm B, Worwa C, Meyers P, geted ventilation is more suitable than pres-
Collaborative Groups; Tarnow-Mordi W, Mammel MC: Bi-level CPAP does not im- sure-limited ventilation for preterm infants:
Stenson B, Kirby A, Juszczak E, Donoghoe M, prove gas exchange when compared with a systematic review and meta-analysis. Arch
Deshpande S, et al: Outcomes of two trials of conventional CPAP for the treatment of neo- Dis Child Fetal Neonatal Ed 2014; 99:F158
oxygen-saturation targets in preterm infants. nates recovering from respiratory distress F165.
N Engl J Med 2016;374:749760. syndrome. Arch Dis Child Fetal Neonatal Ed 97 Wheeler K, Klingenberg C, McCallion N,
72 Saugstad OD, Aune D: Optimal oxygenation 2015;100:F31F34. Morley CJ, Davis PG: Volume-targeted ver-
of extremely low birth weight infants: a meta- 85 Lemyre B, Davis PG, de Paoli AG, Kirpalani sus pressure-limited ventilation in the neo-
analysis and systematic review of the oxygen H: Nasal intermittent positive pressure venti- nate. Cochrane Database Syst Rev 2010;
saturation target studies. Neonatology 2014; lation (NIPPV) versus nasal continuous posi- 11:CD003666.
105: 5563. tive airway pressure (NCPAP) for preterm 98 Keszler M, Nassabeh-Montazami S, Abuba-
73 Stenson BJ: Oxygen saturation targets for ex- neonates after extubation. Cochrane Data- kar K: Evolution of tidal volume require-
tremely preterm infants after the NeOProM base Syst Rev 2014;9:CD003212. ment during the first 3 weeks of life in infants
trials. Neonatology 2016;109:352358. 86 Dumpa V, Katz K, Northrup V, Bhandari V: <800 g ventilated with volume guarantee.
74 Sola A, Golombek SG, Montes Bueno MT, SNIPPV versus NIPPV: does synchronization Arch Dis Child Fetal Neonatal Ed 2009;
Lemus-Varela L, Zuluaga C, Domnguez F, et matter? J Perinatol 2012;32:438442. 94:F279F282.
al: Safe oxygen saturation targeting and mon- 87 Bancalari E, Claure N: The evidence for non- 99 Rimensberger PC, Cox PN, Frndova H, Bry-
itoring in preterm infants: can we avoid hy- invasive ventilation in the preterm infant. an AC: The open lung during small tidal vol-
poxia and hyperoxia? Acta Paediatr 2014;103: Arch Dis Child Fetal Neonatal Ed 2013; ume ventilation: concepts of recruitment
10091018. 98:F98F102. and optimal positive end-expiratory pres-
75 Van Zanten HA, Tan RN, van den Hoogen A, 88 Kirpalani H, Millar D, Lemyre B, Yoder BA, sure. Crit Care Med 1999;27:19461952.
Lopriore E, te Pas AB: Compliance in oxygen Chiu A, Roberts RS; NIPPV Study Group: A 100 De Jaegere A, van Veenendaal MB, Michiels
saturation targeting in preterm infants: a sys- trial comparing noninvasive ventilation strat- A, van Kaam AH: Lung recruitment using
tematic review. Eur J Pediatr 2015;174:1561 egies in preterm infants. N Engl J Med 2013; oxygenation during open lung high-fre-
1572. 369:611620. quency ventilation in preterm infants. Am J
76 Lim K, Wheeler KI, Gale TJ, Jackson HD, 89 Millar D, Lemyre B, Kirpalani H, Chiu A, Yo- Respir Crit Care Med 2006;174:639645.
Kihlstrand JF, Sand C, et al: Oxygen satura- der BA, Roberts RS: A comparison of bilevel 101 Cools F, Offringa M, Askie LM: Elective high
tion targeting in preterm infants receiving and ventilator-delivered non-invasive respi- frequency oscillatory ventilation versus con-
continuous positive airway pressure. J Pediatr ratory support. Arch Dis Child Fetal Neonatal ventional ventilation for acute pulmonary
2014;164:730736. Ed 2016;101:2125. dysfunction in preterm infants. Cochrane
77 Poets CF, Roberts RS, Schmidt B, Whyte RK, 90 Wilkinson D, Andersen C, ODonnell CP, de Database Syst Rev 2015;3:CD000104.
Asztalos EV, Bader D, et al: Association be- Paoli AG, Manley BJ: High flow nasal cannula 102 Zivanovic S, Peacock J, Alcazar-Paris M, Lo
tween intermittent hypoxemia or bradycardia for respiratory support in preterm infants. JW, Lunt A, Marlow N, Calvert S, Gree-
and late death or disability in extremely pre- Cochrane Database Syst Rev 2016;2:CD006405. nough A; United Kingdom Oscillation
term infants. JAMA 2015;314:595603. 91 Reynolds P, Leontiadi S, Lawson T, Otunla T, Study Group: Late outcomes of a random-
78 Van Kaam AH, Hummler HD, Wilinska M, Ejiwumi O, Holland N: Stabilisation of pre- ized trial of high-frequency oscillation in
Swietlinski J, Lal MK, te Pas AB, et al: Auto- mature infants in the delivery room with nasal neonates. N Engl J Med 2014; 370: 1121
mated versus manual oxygen control with dif- high flow. Arch Dis Child Fetal Neonatal Ed 1130.
ferent saturation targets and modes of respi- 2016;101:F284F287. 103 Manley BJ, Doyle LW, Owen LS, Davis PG:
ratory support in preterm infants. J Pediatr 92 Roberts CT, Owen LS, Manley BJ, Donath Extubating extremely preterm infants: pre-
2015;167:545550. SM, Davis PG: A multicentre, randomised dictors of success and outcomes following
79 Davis PG, Henderson-Smart DJ: Nasal con- controlled, non-inferiority trial, comparing failure. J Pediatr 2016;173:4549.
tinuous positive airway pressure immediately high flow therapy with nasal continuous pos- 104 Danan C, Durrmeyer X, Brochard L, Dec-
after extubation for preventing morbidity in itive airway pressure as primary support for obert F, Benani M, Dassieu G: A random-
preterm infants. Cochrane Database Syst Rev preterm infants with respiratory distress (the ized trial of delayed extubation for the re-
2003;2:CD000143. HIPSTER trial): study protocol. BMJ Open duction of reintubation in extremely pre-
80 Rojas-Reyes MX, Morley CJ, Soll R: Prophy- 2015;5:e008483. term infants. Pediatr Pulmonol 2008; 43:
lactic versus selective use of surfactant in pre- 93 SUPPORT Study Group of the Eunice Kennedy 117124.
venting morbidity and mortality in preterm Shriver NICHD Neonatal Research Network; 105 Buzzella B, Claure N, DUgard C, Bancalari
infants. Cochrane Database Syst Rev 2012; Finer NN, Carlo WA, Walsh MC, Rich W, E: A randomized controlled trial of two nasal
3:CD000510. Gantz MG, Laptook AR, Yoder BA, et al: Early continuous positive airway pressure levels
81 Davis PG, Morley CJ, Owen LS: Non-invasive CPAP versus surfactant in extremely preterm after extubation in preterm infants. J Pediatr
respiratory support of preterm neonates with infants. N Engl J Med 2010;362:19701979. 2014;164:4651.
158.232.240.113 - 9/26/2016 8:31:19 PM

European Consensus Guidelines on the Neonatology 2017;111:107125 123


Management of RDS 2016 Update DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
106 Schmidt B, Roberts RS, Davis P, Doyle LW, 118 https://www.npeu.ox.ac.uk/minidex. dence-based review. Semin Perinatol 2014;
Barrington KJ, Ohlsson A, et al; Caffeine for 119 Baud O, Maury L, Lebail F, Ramful D, El 38:1724.
Apnea of Prematurity Trial Group: Caffeine Moussawi F, Nicaise C, et al; PREMILOC 133 Trivedi A, Sinn JKH: Early versus late ad-
therapy for apnea of prematurity. N Engl J Trial Study Group: Effect of early low-dose ministration of amino acids in preterm in-
Med 2006;354:21122121. hydrocortisone on survival without bron- fants receiving parenteral nutrition. Coch-
107 Schmidt B, Roberts RS, Davis P, Doyle LW, chopulmonary dysplasia in extremely pre- rane Database Syst Rev 2013;7:CD008771.
Barrington KJ, Ohlsson A, et al; Caffeine for term infants (PREMILOC): a double-blind, 134 Moyses HE, Johnson MJ, Leaf AA, Cornelius
Apnea of Prematurity Trial Group: Long- placebo-controlled, multicentre, random- VR: Early parenteral nutrition and growth
term effects of caffeine therapy for apnea of ised trial. Lancet 2016;387:18271836. outcomes in preterm infants: a systematic
prematurity. N Engl J Med 2007; 357: 1893 120 Bassler D, Plavka R, Shinwell ES, Hallman review and meta-analysis. Am J Clin Nutr
1902. M, Jarreau PH, Carnielli V, et al; NEUROSIS 2013;97:816826.
108 Dobson NR, Patel RM, Smith PB, Kuehn Trial Group: Early inhaled budesonide for 135 Bonsante F, Iacobelli S, Latorre G, Rigo J, De
DR, Clark J, Vyas-Read S, et al: Trends in the prevention of bronchopulmonary dys- Felice C, Robillard PY, et al: Initial amino
caffeine use and association between clinical plasia. N Engl J Med 2015;373:14971506. acid intake influences phosphorus and cal-
outcomes and timing of therapy in very low 121 Yeh TF, Chen CM, Wu SY, Husan Z, Li TC, cium homeostasis in preterm infants it is
birth weight infants. J Pediatr 2014;164:992 Hsieh WS, et al: Intratracheal administra- time to change the composition of the early
998. tion of budesonide/surfactant to prevent parenteral nutrition. PLoS One 2013; 8:
109 Taha D, Kirkby S, Nawab U, Dysart KC, bronchopulmonary dysplasia. Am J Respir e72880.
Genen L, Greenspan JS, Aghai ZH: Early caf- Crit Care Med 2016;193:8695. 136 Vlaardingerbroek H, Vermeulen MJ, Rook
feine therapy for prevention of bronchopul- 122 Phillipos E, Solevg AL, Pichler G, Aziz K, D, van den Akker CH, Dorst K, Wattimena
monary dysplasia in preterm infants. J Ma- van Os S, OReilly M, et al: Heart rate assess- JL, et al: Safety and efficacy of early paren-
tern Fetal Neonatal Med 2014; 27: 1698 ment immediately after birth. Neonatology teral lipid and high-dose amino acid admin-
1702. 2016;109:130138. istration to very low birth weight infants. J
110 Lodha A, Seshia M, McMillan DD, Bar- 123 Bruschettini M, Romantsik O, Zappettini S, Pediatr 2013;163:638644.
rington K, Yang J, Lee SK, Shah PS; Cana- Ramenghi LA, Calevo MG: Transcutaneous 137 Morgan J, Bombell S, McGuire W: Early tro-
dian Neonatal Network: Association of early carbon dioxide monitoring for the prevention phic feeding versus enteral fasting for very pre-
caffeine administration and neonatal out- of neonatal morbidity and mortality. Coch- term or very low birth weight infants. Coch-
comes in very preterm neonates. JAMA Pe- rane Database Syst Rev 2016;2:CD011494. rane Database Syst Rev 2013;3: CD000504.
diatr 2015;169:3338. 124 Hyttel-Sorensen S, Pellicer A, Alderliesten 138 Morgan J, Young L, McGuire W: Delayed
111 Steer P, Flenady V, Shearman A, Charles B, T, Austin T, van Bel F, Benders M, et al: Ce- introduction of progressive enteral feeds to
Gray PH, Henderson-Smart D, et al: High rebral near infrared spectroscopy oximetry prevent necrotising enterocolitis in very low
dose caffeine citrate for extubation of pre- in extremely preterm infants: phase II ran- birth weight infants. Cochrane Database
term infants: a randomised controlled trial. domised clinical trial. BMJ 2015;350:g7635. Syst Rev 2014;12:CD001970.
Arch Dis Child Fetal Neonatal Ed 2004; 125 Reilly MC, Vohra S, Rac VE, Dunn M, Fer- 139 Morgan J, Young L, McGuire W: Slow ad-
89:F499F503. relli K, Kiss A, et al: Randomized trial of oc- vancement of enteral feed volumes to pre-
112 Mohammed S, Nour I, Shabaan AE, Shou- clusive wrap for heat loss prevention in pre- vent necrotising enterocolitis in very low
man B, Abdel-Hady H, Nasef N: High versus term infants. J Pediatr 2015;166:262268. birth weight infants. Cochrane Database
low-dose caffeine for apnea of prematurity: 126 McCarthy LK, Molloy EJ, Twomey AR, Syst Rev 2015;10:CD001241.
a randomized controlled trial. Eur J Pediatr Murphy JF, ODonnell CP: A randomized 140 Quigley M, McGuire W: Formula versus do-
2015;174:949956. trial of exothermic mattresses for preterm nor breast milk for feeding preterm or low
113 Woodgate PG, Davies MW: Permissive hy- newborns in polyethylene bags. Pediatrics birth weight infants. Cochrane Database
percapnia for the prevention of morbidity 2013;132:e135e141. Syst Rev 2014;4:CD002971.
and mortality in mechanically ventilated 127 Meyer MP, Hou D, Ishrar NN, Dito I, te Pas 141 Polin RA, Watterberg K, Benitz W, Eichen-
newborn infants. Cochrane Database Syst AB: Initial respiratory support with cold, dry wald E: The conundrum of early-onset sep-
Rev 2001;2:CD002061. gas versus heated humidified gas and admis- sis. Pediatrics 2014;133:11221123.
114 Thome UH, Genzel-Boroviczeny O, sion temperature of preterm infants. J Pedi- 142 Soll RF, Edwards WH: Antibiotic use in neo-
Bohnhorst B, Schmid M, Fuchs H, Rohde O, atr 2015;166:245250. natal intensive care. Pediatrics 2015; 135:
et al; PHELBI Study Group: Permissive hy- 128 Sinclair JC: Servo-control for maintaining 928929.
percapnia in extremely low birthweight in- abdominal skin temperature at 36 C in low
143 Cotton CM: Antibiotic stewardship: reas-
fants (PHELBI): a randomised controlled birth weight infants. Cochrane Database sessment of guidelines for management of
multicentre trial. Lancet Respir Med 2015;3: Syst Rev 2002;1: D001074. neonatal sepsis. Clin Perinatol 2015;42:195
534543. 129 Conde-Agudelo A, Daz-Rossello JL: Kanga- 206.
115 Doyle LW, Ehrenkranz RA, Halliday HL: Late roo mother care to reduce morbidity and 144 www.nice.org.uk/guidance/cg149.
(>7 days) postnatal corticosteroids for chron- mortality in low birthweight infants. Coch- 145 Faust K, Hrtel C, Preuss M, Rabe H, Roll C,
ic lung disease in preterm infants. Cochrane rane Database Syst Rev 2014;4:CD002771. Emeis M, et al; Neocirculation Project and
Database Syst Rev 2014;5:CD001145. 130 Cho ES, Kim SJ, Kwon MS, Cho H, Kim EH, the German Neonatal Network (GNN):
116 Doyle LW, Halliday HL, Ehrenkranz RA, Jun EM, Lee S: The effects of kangaroo care Short-term outcome of very-low-birth-
Davis PG, Sinclair JC: An update on the im- in the neonatal intensive care unit on the weight infants with arterial hypotension in
pact of postnatal systemic corticosteroids on physiological functions of preterm infants, the first 24 h of life. Arch Dis Child Fetal
mortality and cerebral palsy in preterm in- maternal-infant attachment, and maternal Neonatal Ed 2015;100:F388F392.
fants: effect modification by risk of broncho- stress. J Pediatr Nurs 2016;31:430438. 146 Batton B, Li L, Newman NS, Das A, Watter-
pulmonary dysplasia. J Pediatr 2014; 165: 131 Bell EF, Acarregui MJ: Restricted versus lib- berg KL, Yoder BA, et al; Eunice Kennedy
12581260. eral water intake for preventing morbidity Shriver National Institute of Child Health
117 Jefferies AL: Postnatal corticosteroids to and mortality in preterm infants. Cochrane and Human Development Neonatal Re-
treat or prevent chronic lung disease in pre- Database Syst Rev 2014;12:CD000503. search Network: Evolving blood pressure
term infants. Paediatr Child Health 2012;17: 132 Barrington KJ: Management during the first dynamics for extremely preterm infants. J
573574. 72 h of age of the periviable infant: an evi- Perinatol 2014;34:301305.
158.232.240.113 - 9/26/2016 8:31:19 PM

124 Neonatology 2017;111:107125 Sweet etal.


DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:
147 Batton B, Li L, Newman NS, Das A, Watter- 156 https://www.nichd.nih.gov/about/Docu- 167 Stevens B, Yamada J, Lee GY, Ohlsson A: Su-
berg KL, Yoder BA, et al; Eunice Kennedy ments/TOP_Protocol.pdf. crose for analgesia in newborn infants un-
Shriver National Institute of Child Health 157 Weisz DE, More K, McNamara PJ, Shah PS: dergoing painful procedures. Cochrane Da-
and Human Development Neonatal Re- PDA ligation and health outcomes: a meta- tabase Syst Rev 2013;1:CD001069.
search Network: Use of antihypotensive analysis. Pediatrics 2014;133:e1024e1046. 168 Tan K, Lai NM, Sharma A: Surfactant for
therapies in extremely preterm infants. Pe- 158 Heuchan AM, Clyman RI: Managing the bacterial pneumonia in late preterm and
diatrics 2013;131:e1865e1873. patent ductus arteriosus: current treatment term infants. Cochrane Database Syst Rev
148 Roehr CC, te Pas AB, Dold SK, Breindahl M, options. Arch Dis Child Fetal Neonatal Ed 2012;2:CD008155.
Blennow M, Rdiger M, Gupta S: Investigat- 2014;99:F431F436. 169 Vento G, Tana M, Tirone C, Aurilia C, Lio
ing the European perspective of neonatal 159 Ohlsson A, Walia R, Shah SS: Ibuprofen for A, Perelli S, Ricci C, Romagnoli C: Effective-
point-of-care echocardiography in the neo- the treatment of patent ductus arteriosus in ness of treatment with surfactant in prema-
natal intensive care unit a pilot study. Eur preterm or low birth weight (or both) in- ture infants with respiratory failure and pul-
J Pediatr 2013;172:907911. fants. Cochrane Database Syst Rev 2015; monary infection. Acta Biomed 2012; 83
149 Subhedar NV, Shaw NJ: Dopamine versus 2:CD003481. (suppl 1):3336.
dobutamine for hypotensive preterm in- 160 Ohlsson A, Shah PS: Paracetamol (acet- 170 Aziz A, Ohlsson A: Surfactant for pulmo-
fants. Cochrane Database Syst Rev 2003; aminophen) for patent ductus arteriosus in nary haemorrhage in neonates. Cochrane
3:CD001242. preterm or low-birth-weight infants. Coch- Database Syst Rev 2012;7:CD005254.
150 Ruoss JL, McPherson C, DiNardo J: Inotro- rane Database Syst Rev 2015;3:CD010061. 171 Bozda , Dilli D, Gkmen T, Dilmen U:
pe and vasopressor support in neonates. 161 Kluckow M, Jeffery M, Gill A, Evans N: A Comparison of two natural surfactants for
Neoreviews 2015;16:e351e361. randomised placebo-controlled trial of early pulmonary hemorrhage in very low-birth-
151 Dempsey EM, Barrington KJ, Marlow N, treatment of the patent ductus arteriosus. weight infants: a randomized controlled tri-
ODonnell CP, Miletin J, Naulaers G, et al; Arch Dis Child Fetal Neonatal Ed 2014; al. Am J Perinatol 2015;32:211218.
HIP Consortium: Management of Hypoten- 99:F99F104. 172 Askie LM, Ballard RA, Cutter GR, Dani C,
sion in Preterm infants (The HIP Trial): a 162 https://www.npeu.ox.ac.uk/baby-oscar/ Elbourne D, Field D, Hascoet JM, et al; Me-
randomised controlled trial of hypotension protocol. ta-Analysis of Preterm Patients on Inhaled
management in extremely low gestational 163 Hummel P, Lawlor-Klean P, Weiss MG: Va- Nitric Oxide Collaboration: Inhaled nitric
age newborns. Neonatology 2014; 105: 275 lidity and reliability of the N-PASS assess- oxide in preterm infants: an individual-pa-
281. ment tool with acute pain. J Perinatol 2010; tient data meta-analysis of randomized tri-
152 Paradisis M, Osborn DA: Adrenaline for 30:474478. als. Pediatrics 2011;128:729739.
prevention of morbidity and mortality in 164 Durrmeyer X, Dahan S, Delorme P, Blary S, 173 Breatnach CR, Flanagan F, James A, Corco-
preterm infants with cardiovascular com- Dassieu G, Caeymaex L, Carbajal R: Assess- ran JD, Franklin O, El-Khuffash A: The use
promise. Cochrane Database Syst Rev 2004; ment of atropine-sufentanil-atracurium an- of inhaled nitric oxide in a tertiary neonatal
1:CD003958. aesthesia for endotracheal intubation: an ob- intensive care unit. Ir Med J 2015; 108: 275
153 Ibrahim H, Sinha IP, Subhedar NV: Cortico- servational study in very premature infants. 278.
steroids for treating hypotension in preterm BMC Pediatr 2014;14:120. 174 Ellsworth MA, Harris MN, Carey WA,
infants. Cochrane Database Syst Rev 2011; 165 Dekker J, Lopriore E, Rijken M, Rijntjes-Ja- Spitzer AR, Clark RH: Off-label use of in-
12:CD003662. cobs E, Smits-Wintjens V, te Pas A: Sedation haled nitric oxide after release of NIH con-
154 Whyte R, Kirpalani H: Low versus high hae- during minimally invasive surfactant thera- sensus statement. Pediatrics 2015; 135: 643
moglobin concentration threshold for blood py in preterm infants. Neonatology 2016; 648.
transfusion for preventing morbidity and 109:308313. 175 Shah DM, Kluckow M: Early functional
mortality in very low birth weight infants. 166 Bell R, de Waal K, Zanini R: Opioids for echocardiogram and inhaled nitric oxide:
Cochrane Database Syst Rev 2011; 11: neonates receiving mechanical ventilation: a usefulness in managing neonates born fol-
CD000512. systematic review and meta-analysis. Arch lowing extreme preterm premature rupture
155 Whyte RK, Kirpalani H, Asztalos EV, An- Dis Child Fetal Neonatal Ed 2010; 95:F241 of membranes (PPROM). J Paediatr Child
dersen C, Blajchman M, Heddle N, et al; F251. Health 2011;47:340345.
PINTOS Study Group: Neurodevelopmen- 176 Cheng DR, Peart S, Tan K, Sehgal A: Nitric
tal outcome of extremely low birth weight therapy in preterm infants: rationalised ap-
infants randomly assigned to restrictive or proach based on functional neonatal echo-
liberal hemoglobin thresholds for blood cardiography. Acta Paediatr 2016; 105: 165
transfusion. Pediatrics 2009;123:207213. 171.

158.232.240.113 - 9/26/2016 8:31:19 PM

European Consensus Guidelines on the Neonatology 2017;111:107125 125


Management of RDS 2016 Update DOI: 10.1159/000448985
HINARI Guatemala
Downloaded by:

Você também pode gostar