You are on page 1of 7

Clinical Nutrition 42 (2023) 22e28

Contents lists available at ScienceDirect

Clinical Nutrition
journal homepage: http://www.elsevier.com/locate/clnu

Randomized Control Trials

Effect of arachidonic and docosahexaenoic acid supplementation on


respiratory outcomes and neonatal morbidities in preterm infants
Kristina Wendel a, c, *, Marlen Fossan Aas a, c, Gunnthorunn Gunnarsdottir b, c,
Madelaine Eloranta Rossholt a, d, Marianne Bratlie a, d, Tone Nordvik a, c,
Erlend Christoffer Sommer Landsend e, Drude Fugelseth a, c, Magnus Domello € f f,
g a, c a, c
Are Hugo Pripp , Tom Stiris , Sissel Jennifer Moltu
a
Department of Neonatal Intensive Care, Oslo University Hospital, Norway
b
Department of Pediatric Neurology, Oslo University Hospital, Norway
c
Institute of Clinical Medicine, University of Oslo, Norway
d
Department of Pediatrics and Adolescence Medicine, Oslo University Hospital, Norway
e
Department of Ophthalmology, Oslo University Hospital, Norway
f
Department of Clinical Sciences, Pediatrics, Umea University, Sweden
g
Oslo Centre of Biostatistics and Epidemiology, Oslo University Hospital, Norway

a r t i c l e i n f o s u m m a r y

Article history: Background & aims: Studies have suggested that supplementation with docosahexaenoic acid (DHA) to
Received 31 May 2022 preterm infants might be associated with an increased risk of bronchopulmonary dysplasia (BPD). Our
Accepted 11 November 2022 aim was to investigate the effect of enteral supplementation with arachidonic acid (ARA) and DHA on
short-term respiratory outcomes and neonatal morbidities in very preterm infants.
Keywords: Methods: This is a secondary analysis of data from the ImNuT (Immature, Nutrition Therapy) study, a
Preterm
randomized double blind clinical trial. Infants with gestational age less than 29 weeks were randomized
Fatty acid supplementation
to receive a daily enteral supplement with ARA 100 mg/kg and DHA 50 mg/kg (intervention) or medium
Arachidonic acid
Docosahexaenoic acid
chain triglycerides (MCT) oil (control), from second day of life to 36 weeks postmenstrual age. Study
Bronchopulmonary dysplasia outcomes included duration of respiratory support, incidence of BPD and other major morbidities
Neonatal morbidities associated with preterm birth.
Results: 120 infants with mean (SD) gestational age 26.4 (1.7) weeks were randomized and allocated to
either the intervention or control group. Supplementation with ARA and DHA led to a significant
reduction in number of days with respiratory support (mean (95% CI) 63.4 (56.6e71.3) vs 80.6 (72.4
e88.8); p ¼ 0.03) and a lower oxygen demand (FiO2) (mean (95% CI) 0.26 (0.25e0.28) vs 0.29 (0.27
e0.30); p ¼ 0.03) compared to control treatment. There were no clinically important differences in
incidence of BPD and other major morbidities between the treatment groups.
Conclusions: Supplementation with ARA and DHA to preterm infants was safe and might have a bene-
ficial effect on respiratory outcomes.
Clinical trial registration: The trial has been registered in www.clinicaltrials.gov, ID: NCT03555019.
© 2022 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).

Abbreviations: ARA, Arachidonic acid; BPD, Bronchopulmonary dysplasia; CPAP, continuous positive airway pressure; CRIB, Clinical Risk Index for Babies; DHA, Doco-
sahexaenoic acid; DMC, Data Monitoring Committee; FA, Fatty acid; GA, Gestational age; HR, Hazard ratio; IVH, Intraventricular hemorrhage; LCPUFA, Long-chain poly-
unsaturated fatty acid; LOS, Late-onset septicemia; MRI, Magnetic Resonance Imaging; MCT, Medium chain triglycerides; MD, Mean difference; NEC, Necrotizing
enterocolitis; PDA, Patent ductus arteriosus; PMA, Postmenstrual age; PVL, Periventricular leukomalacia; ROP, Retinopathy of prematurity; RR, Respiratory rate or Risk ratio;
TPTEF/Te ratio, Time to peak tidal expiratory flow as a ratio of expiratory time; Vt, Tidal volume.
* Corresponding author. Department of Neonatal Intensive Care, Oslo University Hospital, Postboks 4950 Nydalen, 0424 Oslo, Norway.
E-mail addresses: krwend@ous-hf.no, kristinalawendel@gmail.com (K. Wendel).

https://doi.org/10.1016/j.clnu.2022.11.012
0261-5614/© 2022 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en enero 18, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
K. Wendel, M.F. Aas, G. Gunnarsdottir et al. Clinical Nutrition 42 (2023) 22e28

1. Introduction published [17]. The primary outcome in the ImNuT study was to
determine the effect of ARA and DHA supplementation on brain
Bronchopulmonary dysplasia (BPD) is a common complication growth and maturation in very preterm infants. Pre-specified sec-
of preterm birth, affecting more than 50% of infants born before 28 ondary outcomes included respiratory outcomes, incidence of
weeks gestational age (GA) [1]. BPD is mainly caused by arrested neonatal morbidities, quality of growth, cardiovascular health and
pulmonary development and sustained postnatal inflammation neuro-development. The Norwegian Regional Ethics Committee
aggravated by mechanical ventilation and oxidative stress [2,3]. approved the study (2016-003700-31) and a data monitoring
Approaches to reduce postnatal inflammation are numerous, but committee (DMC) with access to reported adverse effects and
the incidence of BPD has remained unchanged over the last decades clinical outcomes reviewed the study for safety and efficacy. We
[4]. The long-chain polyunsaturated fatty acids (LCPUFAs) arach- have followed the Consolidated Standards of Reporting Trials
idonic acid (ARA) and docosahexaenoic acid (DHA) are important (CONSORT) guideline.
structural lipids in the cell membrane, but they also act as signal
molecules attenuating the inflammatory cascade [5]. ARA and DHA 2.2. Study participants
mainly influence the inflammatory process through the formation
of specialized pro-resolving lipid mediators; lipoxins derived from Infants born at Oslo University Hospital with GA less than 29
ARA and protectins, maresins and d-series resolvins derived from weeks were eligible to participate in the study. Exclusion criteria
DHA [6,7]. DHA has additional immunomodulatory properties, were congenital malformations, chromosomal abnormalities or
including an inhibiting effect on the production of pro- critical illness with short life expectancy. Study participation
inflammatory cytokines [8]. Fetal plasma concentrations of ARA required written informed, parental consent within 48 h after
are physiologically high at the beginning of the third trimester, with birth.
an ARA:DHA ratio around 3:1 [9]. Infants born before 29 weeks GA
are deprived of the fetal accretion of LCPUFAs occurring during the
third trimester and rapidly become deficient in ARA and DHA after 2.3. Randomization and blinding
birth [10]. Postnatally, preterm infants rely on exogenous supply of
ARA and DHA since their capacity of endogenous synthesis from We used a computer-generated list of random numbers for
linoleic acid and alfa-linolenic acid is low [11]. Human breast milk allocation of the participants to one of the two treatment groups.
provides ARA and DHA in a 2:1 ratio, but in insufficient amounts to The block size was 4, 6 or 8, randomly alternated. To decrease the
prevent a postnatal deficiency [12]. risk of unbalanced baseline characteristics, we stratified the
Low postnatal DHA concentrations are associated with an randomization by growth status at birth (small for gestational age
increased risk of BPD [10]. However, studies on the effect of DHA or not) with an allocation ratio of 1:1 within each block. In case of
supplementation on respiratory outcomes in preterm infants have multiple births, the infants were allocated to the same treatment by
shown conflicting results and there is a concern that DHA supple- randomizing only the first infant. After randomization, the local
mentation may have adverse effects. Although a high-DHA diet investigator contacted the study pharmacist for preparation of
given to lactating mothers of premature infants reduced the inci- allocated fatty acid intervention. All participants, investigators and
dence of BPD in one study [13], a similar intervention study was staff, except for the pharmacist were blinded to the fatty acid
terminated early due to higher rates of BPD in the intervention assignment.
group [14]. Higher rates of BPD were also reported in a randomized
controlled trial with enteral DHA supplementation to infants less 2.4. Trial intervention
than 29 weeks GA [15]. High doses of DHA without concomitant
ARA supplementation are associated with a significant reduction in Infants randomized to the ARA:DHA group received a fatty
ARA blood concentrations and show less beneficial effects on acid supplement containing ARA and DHA in a 2:1 ratio (For-
developmental outcomes in term infants compared to more mod- mulaid™, DSM Nutritional Products Inc.). Infants randomized to
erate DHA supplementation [16]. This suggests that a balanced the control group received medium chain triglycerides (MCT-
intake of ARA and DHA is important. Few studies have investigated oil™, Nutricia) (product details are found in Supplement,
the effect of both ARA and DHA on respiratory outcomes and Appendix 1). MCT-oil is currently the standard lipid supple-
neonatal morbidities in preterm infants. ment in our department to enhance enteral energy supply and
The objective of ImNuT (Immature, Nutrition Therapy), a does not contain ARA or DHA. Lipid supplementation in a dosage
double blind randomized controlled trial, was to determine the of 0.2 ml/kg was started on the second day of life and advanced
effect of ARA and DHA supplementation on growth, inflamma- to target 0.4 ml/kg from day four and onwards, resulting in an
tion and metabolism in premature infants with a gestational age intake of 100 mg/kg of ARA and 50 mg/kg of DHA in the inter-
less than 29 weeks [17]. This study is a secondary analysis of data vention group. The dose was weekly weight-adjusted, but weight
from the ImNuT trial. We hypothesized that a balanced supple- at birth was used until birth weight was regained. The enteral
mentation of ARA and DHA would improve respiratory outcomes fatty acid supplementation was administered in the feeding tube
and reduce morbidities associated with preterm birth. Further, as a daily bolus until 36 weeks postmenstrual age (PMA). Study
we hypothesized that supplementation with ARA and DHA would participants transferred to local hospitals continued to receive
result in increased LCPUFA blood concentrations during the fatty acid supplementation. All study participants followed a
hospitalization. standardized nutritional protocol to accommodate international
recommendations [18] (Supplement, eTable 1). The intravenous
2. Material & methods solution used for parenteral nutrition (PN) contained a composite
lipid emulsion of soybean and olive oil emulsion (Clinoleic®,
2.1. Study design Baxter). This emulsion has a small amount of ARA, but no DHA.
Full enteral nutrition was defined as  150 mL/kg/day of human
The ImNuT study was a double-blind randomized controlled milk fortified with PreNAN FM 85® (Nestle ) 3 g per100 mL. The
trial conducted at a tertiary neonatal intensive care unit at Oslo human milk fortifier contained 1.6 mg DHA per gram and no
University Hospital in Norway. The trial design has previously been ARA.
23

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en enero 18, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
K. Wendel, M.F. Aas, G. Gunnarsdottir et al. Clinical Nutrition 42 (2023) 22e28

2.5. Study outcomes 2.6. Statistical methods

We registered respiratory outcomes from birth up to 40 weeks The sample size calculation was based on the primary
PMA, including number of days requiring respiratory support outcome in the ImNuT trial; 120 infants would provide 80% po-
(mechanical ventilation, continuous positive airway pressure wer to detect a 0.04 difference in mean diffusivity (mm2/sec) in
(CPAP), high flow nasal cannula 3 L/min or oxygen therapy), major white matter tracts on magnetic resonance imaging (MRI)
mean oxygen demand (FiO2) during respiratory support, and use at term equivalent age. We performed efficacy analyses on the
of postnatal steroids. BPD was defined by need of respiratory intention-to-treat (ITT) population and for pre-specified study
support or oxygen demand at 36 weeks PMA [19]. The target range outcomes on infants who survived up to 36 weeks PMA. Cumu-
for oxygen saturation during oxygen supplementation was lative days of respiratory support was described by using the
90e94%. Kaplan-Meyer technique and tested with a log-rank test. Dropout
Lung function was assessed at 36 ± 2 weeks PMA by a tidal and death were considered as non-events and censored at the
breathing flow-volume test [20]. The test procedure is described in time of discontinuation. For analysis of days with mechanical
supplement (Appendix 2). Tidal breathing parameters included in ventilation, we used quantile regression since the data was not
the analysis were respiratory rate (RR), tidal volume (Vt)/kg, and normally distributed. Other normally distributed continuous
time to peak tidal expiratory flow as a ratio of expiratory time outcomes were analyzed by an independent t-test. The effect size
(TPTEF/Te ratio). Infants who needed respiratory support or were for dichotomous outcomes was estimated by risk ratio using a
transferred to local hospitals before the time of evaluation were chi-square test or Fishers exact test as appropriate. For the
excluded from the lung function test. analysis of ARA and DHA blood concentrations, we used mixed
We also assessed the occurrence of neonatal morbidities models for repeated measures with a subject-specific random
including retinopathy of prematurity (ROP), necrotizing enteroco- intercept. Day 3 was used as baseline, whereas fixed effects were
litis (NEC), intraventricular hemorrhage (IVH), patent ductus arte- fatty acid blood concentrations at day 3, treatment group, time
riosus (PDA) requiring treatment, and late-onset septicemia (LOS). and the interaction between group and time. All results are
For detailed definitions, see supplement (Appendix 3). Major presented un-adjusted. To adjust for baseline imbalances, a post-
morbidity was defined as the presence of at least one of the hoc sensitivity analysis was performed using cox regression and
following morbidities: any BPD, severe brain injury (IVH grade 3 multiple regression. For all analyses, we considered a p-value of
or periventricular leukomalacia (PVL)) or ROP stage 3 [21]. <0.05 as statistically significant. SPSS version 26.0 and STATA
We registered the total intake (mg/kg/d) of ARA and DHA from statistical software were used in the statistical analyses.
additional sources (including intravenous PN, human milk, fortifier
and formula) up to 36 weeks PMA [22]. To determine postnatal 3. Results
blood concentrations of ARA and DHA, we collected whole blood
samples on filter sticks (10 mL) at inclusion and then at day 3, 7, 14, 3.1. Study participants
21, 28, and week 36 PMA. ARA and DHA concentrations were
measured as separate fatty acids in dry blood spots; the analytic From April 2018 to January 2021, a total of 121 infants born before
method is described in supplement (Appendix 4). GA 29 weeks were randomized. Sixty-one infants were allocated to

Fig. 1. Study flow diagram.

24

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en enero 18, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
K. Wendel, M.F. Aas, G. Gunnarsdottir et al. Clinical Nutrition 42 (2023) 22e28

the control group and 60 infants to the ARA:DHA group. One patient 3.2. Respiratory outcomes
randomized to the control group was later diagnosed with chro-
mosomal abnormality and therefore excluded from the trial. The ITT Infants in the ARA:DHA group had significantly fewer days with
population included 120 infants (Fig. 1). Neonatal outcome data at 36 respiratory support (63.4 vs 80.6 days; p ¼ 0.03) and a lower mean
weeks PMA was available for 110 of the study participants. Ninety oxygen demand (FiO2 0.27 vs. 0.29; p ¼ 0.03), compared to the
four percent of ordered fatty acid doses were given to the ARA:DHA control group. There was no significant differences in incidence of
group and 92.6% to the control group. One patient in the ARA:DHA BPD, need of respiratory support after 40 weeks PMA or use of
group received 38% of planned doses due to complicated NEC. postnatal steroids. The tidal breathing test at 36 weeks PMA
Baseline characteristics of the study population were similar in the showed that infants in the ARA:DHA group had significantly higher
treatment groups, except for number of infants who had received tidal volumes than the control group (6.26 vs. 5.17 ml/kg; p ¼ 0.02),
two doses of antenatal steroids and Apgar score at 5 min (Table 1). but respiratory rate and TPTEF/Te ratio did not differ (Table 2).
Adjustments for baseline imbalances (number of doses of antenatal
steroids and 5 min Apgar score) in post-hoc sensitivity analysis,
Table 1 resulted in no longer significant differences in days with respiratory
Baseline characteristics (Intention-to-Treat Population). support (hazard ratio (95% CI); 0.72 (0.46e1.13), p ¼ 0.15) and ox-
ygen demand (FiO2 mean difference (MD) (95% CI); 0.016 (0.007-
Control ARA:DHA
(n ¼ 60) (n ¼ 60) 0.40), p ¼ 0.17) between the treatment groups. The difference in
tidal volume remained significant after adjustments (MD (95% CI)
Mothers
Antenatal glucocorticoids any dose, n 60 (100) 60 (100)
ml/kg; 0.99 (0.05e1.93), p ¼ 0.04).
(%)
Antenatal glucocorticoids 2 doses, n (%) 35 (58.3) 48 (80.0)
Cesarean delivery, n (%) 38 (63.3) 29 (48.3) 3.3. Neonatal morbidities
Infants
Gestational age, weeks There were no clinically important differences in incidence of
Mean (SD) 26.2 (1.6) 26.6 (1.7) ROP, NEC, IVH, PDA requiring treatment, or late-onset-septicemia
Median (range) 26.7 (23.0e28.9) 27.0 (22.9e28.9)
Birth weight, g
between the study groups. There was no significant difference in
Mean (SD) 833 (255) 879 (241) number of infants diagnosed with one or more major morbidities,
Median (range) 763 (470e1485) 824 (444e1370) 26 infants in the ARA:DHA group compared to 32 infants in the
Small for gestational age, n (%) 12 (20.0) 11 (18.3) control group. None of the study participants suffered from three
Male sex, n (%) 31 (51.7) 35 (58.3)
major morbidities (Table 3).
Plurality, n (%)
Single 47 (78.3) 44 (73.3)
Twin 10 (16.7) 16 (26.7)
Triplet 3 (5) e
3.4. Intake estimates and blood concentrations of ARA and DHA
Apgar score at 5 min, median (IQR) 7 (6e8) 8 (7e9)
Mechanically ventilated within first 42 (70.0) 35 (58.3) The mean (SD) mg/kg/d intake of ARA was 110.6 (9.4) in the
48 h of life, n (%) ARA:DHA group and 23.8 (1.6) in the control group, while the mean
Surfactant administration, n (%) 56 (93.3) 51 (85.0)
(SD) mg/kg/d intake of DHA was 63.0 (5.7) in the ARA:DHA group
CRIB score, median (IQR) 7 (2e10) 5 (2e9)
Whole blood ARA concentration at 13.0 (3.5) 13.4 (4.5) and 18.4 (2.2) in the control group. The overall ARA concentrations
inclusion mol%, mean (SD) from baseline to 36 weeks PMA did not differ significantly between
Whole blood DHA concentrations at 2.6 (0.8) 2.5 (0.8) the treatment groups, but the DHA blood concentrations was
inclusion mol%, mean (SD) significantly higher in the ARA:DHA group compared to the control
ARA, arachidonic acid; CRIB, Clinical Risk Index for Babies; DHA, docosahexaenoic group with a MD (95% CI) of 0.35 (0.09e0.61) mol%, p ¼ 0.01 at 36
acid; IQR, interquartile range; SD, standard deviation. weeks PMA (Fig. 2).

Table 2
Respiratory outcomes.

Respiratory outcomes Control group (n ¼ 60) ARA:DHA group (n ¼ 60) Comparison: ARA:DHA vs p-value
control group (95%CI)

BPDa, No./total (%) 30/55 (54.5) 23/55 (41.8) RR, 0.77 (0.52e1.14) 0.18
Mild BPD 7/55 (12.7) 6/55 (10.9) RR, 0.86 (0.31e2.39) 0.78
Moderate BPD 14/55 (25.5) 11/55 (20.0) RR, 0.79 (0.39e1.58) 0.49
Severe BPD 9/55 (16.4) 6/55 (10.9) RR, 0.67 (0.25e1.75) 0.41
Days with mechanical ventilation, median (IQR) 8 (1e24) 3 (0e18) MD, 5 (2e12) 0.15
Cumulative days with respiratory supportb, mean (95% CI) 80.6 (72.4e88.8) 63.4 (56.6e71.3) HR, 0.62 (0.40e0.95) 0.03
Oxygen demand (fiO2)c, mean (95% CI) 0.29 (0.27e0.30) 0.26 (0.25e0.28) MD, 0.024 (0.002e0.047) 0.03
Need of respiratory support > 40 weeks PMAd, No./total (%) 17/55 (30.9) 9/55 (16.3) RR, 0.53 (0.26e1.08) 0.07
Postnatal steroids, No./total (%) 28/60 (46.7) 21/60 (35.0) RR, 0.75 (0.48e1.16) 0.19
Tidal breathing parameters at 36 weeks PMAe, No./total (%) 24/60 (40.0) 27/60 (45.0)
Tidal volume (ml/kg), mean (95% CI) 5.17 (4.50e5.83) 6.26 (5.64e6.88) MD, 1.09 (0.21e1.98) 0.02
Respiratory rate (RR/min), mean (95% CI) 77 (70e83) 74 (67e81) MD, 3 (6e13) 0.5
TPTEF/Te ratio, mean (95% CI) 0.35 (0.32e0.39) 0.35 (0.31e0.40) MD, 0.00 (0.05-0.06) 0.93

Abbrevations: ARA, arachidonic acid: DHA, docosahexaenoic acid; BPD, bronchopulmonary dysplasia; PMA, post menstrual age; RR, relative risk; HR, hazard ratio; MD, mean
difference; IQR, interquartile range; TPTEF/Te ratio, time to peak tidal expiratory flow as a ratio of expiratory time.
a
Data are missing for patients who died or withdrawn before 36 weeks PMA.
b
Mechanical ventilation, nCPAP, HFNC 3 L/min or low-flow oxygen.
c
Oxygen demand during need of respiratory support.
d
Data are missing for patients who died or withdrawn before 40 weeks PMA.
e
Data are missing for infants who died, withdrawn, were transferred to local hospitals or needed respiratory support at 36 weeks PMA.

25

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en enero 18, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
K. Wendel, M.F. Aas, G. Gunnarsdottir et al. Clinical Nutrition 42 (2023) 22e28

Table 3
Neonatal morbidities.

Outcomes Control group No./total (%) ARA:DHA group No./total (%) Comparison ARA:DHA vs. p-value
control group Relative risk (95% CI)

ROPa
No ROP 32/55 (58.2) 38/55 (69.1) 1.18 (0.89e1.58) 0.23
Stage  3 7/55 (12.7) 3/55 (5.4) 0.43 (0.12e1.57) 0.32
NEC 2/60 (3.3) 2/60 (3.3) 1.0 (0.37e2.71) 1.0
IVH
No IVH 48/60 (80.0) 47/60 (78.3) 0.98 (0.82e1.18) 0.82
Grade III-IV 4/60 (6.7) 7/60 (11.7) 1.75 (0.54e5.67) 0.53
PDA requiring treatmentb 27/58 (46.6) 24/57 (42.1) 0.90 (0.60e1.36) 0.63
Late-onset-sepsisc 27/57 (47.4) 23/57 (40.4) 0.85 (0.56e1.29) 0.45
Major morbidity in survivors 32/55 (55.2) 26/55 (44.8) 0.81 (0.57e1.16) 0.25
at 36 weeks PMAd

Abbrevations: ROP, retinopathy of prematurity; NEC, necrotizing enterocolitis; IVH, intraventricular hemorrhage; PDA, patent ductus arteriosus; PVL, periventricular leu-
komalacia; PMA, postmenstrual age.
a
Data are missing for infants who died or withdrawn before first ROP-screening.
b
Data are missing for infants who died or withdrawn before first echocardiography at 3e5 days after birth.
c
Infants who died or withdrawn before 72 h were not included in the analysis.
d
Major morbidity included bronchopulmonary dysplasia, severe brain injury (IVH grade III-IV or cystic PVL) or ROP stage 3.

Fig. 2. Postnatal blood concentrations (mol%) of arachidonic acid (ARA) and docosahexaenoic acid (DHA) in the intervention (ARA:DHA) group and control group (Intention-To-
Treat population).

3.5. Safety mainly evaluated the effect of DHA supplementation alone, we


provided a daily dose of fatty acid supplementation containing
There were similar numbers of reported adverse events in the 100 mg/kg ARA and 50 mg/kg DHA. In a large RCT, Collins et al.
ARA:DHA and control group (Supplement, eTable 2). Serious found an increased risk of BPD after enteral supplementation with
adverse events occurred in 13.3% of infants in the ARA:DHA group 60 mg/kg of DHA to infants less than 29 weeks GA, compared to
and 8.3% in the control group (Supplement, eTable 3). A total of five supplementation with a soy emulsion [15]. Similarly, an observa-
study participants died during the study period, 3 (5.0%) in the tional study of 477 infants with birthweight <1250 g reported a
ARA:DHA group and 2 (3.2%) infants in the control group. The in- lower oxygen saturation to FiO2 ratio at 36 weeks PMA after
dividual causes of death are described in Supplement (eTable 4). administration of intravenous lipid emulsions containing DHA
compared to lipid emulsions without DHA [23]. In contrast to these
4. Discussion studies, we found that enteral supplementation with ARA and DHA
in a 2:1 ratio resulted in reduced duration of respiratory support
4.1. Respiratory outcomes and lower oxygen demand during hospitalization. The importance
of a balanced intake of ARA and DHA is supported by the study of
This secondary analysis of a randomized, double-blind Bernhard et al., which found that a low postnatal ARA:DHA ratio in
controlled trial showed that enteral supplementation with ARA blood samples of extremely preterm infants was associated with a
and DHA to preterm infants was safe and possibly associated with higher risk of developing BPD [24]. The possible role of ARA in
improved respiratory outcomes compared to control treatment. preventing lung injury may be explained by its capacity to initiate
While earlier studies on respiratory outcomes in preterm infants resolution of the inflammatory cascade, mainly mediated by ARA-

26

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en enero 18, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
K. Wendel, M.F. Aas, G. Gunnarsdottir et al. Clinical Nutrition 42 (2023) 22e28

derived lipoxins [25]. In a murine model of BPD, injection of Lipoxin et al. demonstrated that feeding term infants with formula con-
A4 led to reduced alveolar simplification and normalized septal taining DHA and not ARA, resulted in lower concentration of ARA in
thickness after exposure to hyperoxia [26]. erythrocytes compared with a control formula without DHA [41].
Dietary supplementation with ARA and DHA in a 2:1 ratio to pre-
4.2. Adjustments of baseline imbalances term baboons improved DHA concentrations but not ARA concen-
trations in brain tissue [42], indicating that ARA concentrations are
After adjustments for baseline imbalances (number of doses of tighter regulated than DHA. This might be explained by the role of
antenatal steroids and 5 min Apgar score), the differences in days ARA as a precursor of potent mediators in intracellular signaling,
with respiratory support and oxygen demand between the treat- implying that the concentration of free ARA must be maintained at
ment groups were no longer significant. Apgar score at 5 min precise levels within the cell [43].
explained most of the variance, while number of doses of antenatal
steroids did not change the validity of the results. The prognostic 4.6. Limitations and strengths
value of Apgar score on neonatal outcomes in extremely preterm
infants has shown conflicting results [27e29]. Of interest, the This study has limitations. The sample size calculation of the
Clinical Risk Index for Babies (CRIB) score, a widely used tool for trial was based on the primary outcome, thus the study was not
prediction of mortality and morbidity in very low birth weight powered to detect differences in respiratory outcomes and
infants [30], did not differ significantly between the ARA:DHA and neonatal morbidities. The relative low number of study participants
control group. affected the robustness of the results. The major strength is that our
study was a double-blind randomized controlled trial, which
4.3. Tidal breathing parameters means that investigators, caregivers and parents were unaware of
treatment assignment.
The tidal breathing test was performed in infants with no need
of respiratory support at 36 ± 2 weeks PMA and consequently did 5. Conclusions
not include infants with moderate and severe BPD. The clinical
importance of the observed difference in tidal volumes between Enteral supplementation with 100 mg/kg ARA and 50 mg/kg
the treatment groups is uncertain, since earlier studies on lung DHA to preterm infants born before 29 weeks of gestation was safe
function in premature infants are contradictory regarding the cor- and may have beneficial effects on short-term respiratory out-
relation between tidal volumes at term and development of chronic comes. Further studies are needed to confirm these findings.
lung disease [31,32].
Funding statement
4.4. Neonatal morbidities
The study was sponsored by funds from the Research Council of
Low postnatal blood concentrations of ARA are not only asso- Norway, with additional contribution from the public foundations
ciated with the development of BPD, but also with an increased risk the South-Eastern Norway Regional Health Authority (Helse Sør-
of late-onset septicemia [10] and severe ROP [33]. Several studies Øst RHF) and Barnestiftelsen, Oslo University Hospital. DSM
have indicated that supplementation with DHA reduced the risk of Nutritional Products Inc. sponsored the investigational nutrition
ROP in premature infants [34e37]. Hellstrom et al. recently product, Formulaid™. This was an investigator-initiated study. The
confirmed this in a multi-center RCT of 207 infants with GA< 28 DSM Nutritional Products Inc. had no role in the design or conduct
weeks. They reported a 50% reduction in severe ROP in infants of the study.
randomized to enteral supplementation with 100 mg/kg ARA and
50 mg/kg DHA, compared to standard treatment [38]. In our study, Author contributions
5.4% in the ARA:DHA group and 12.7% in the control group were
diagnosed with ROP Stage 3. The difference was however not Dr Wendel designed the study, carried out the initial analyses,
statistically significant, probably due to the low number of infants drafted the initial manuscript, and reviewed and revised the
with ROP Stage 3 in the study population. manuscript. Dr Fossan Aas, Dr Gunnarsdottir and Dr Nordvik
included study participants, collected data and reviewed the
4.5. ARA and DHA blood concentrations manuscript. Rossholt and Bratlie collected nutritional data, fol-
lowed up study participants transferred to local hospitals and
The DHA concentrations increased from third week of life in the reviewed the manuscript. Dr Sommer Landsend participated in
ARA:DHA group and became significantly higher compared to the screening of retinopathy of prematurity, collected data and
control group at the end of the study period, but interestingly the reviewed the manuscript. Pripp supervised and conducted statis-
change in ARA concentrations over time did not differ between the tical analysis, and critically reviewed the manuscript. Prof Fugelseth
treatment groups. This is in contrast to two similar RCTs that diagnosed the patency of ductus arteriosus with Doppler echocar-
showed a significant increase in blood concentrations of both ARA diography and critically reviewed the manuscript for important
and DHA after enteral supplementation to extremely preterm in- intellectual content. Prof Domello €f and Prof Stiris made important
fants [38,39]. The knowledge about uptake and distribution of ARA contributions to the study design and critically reviewed the
and DHA in extremely preterm infants is scarce. Multiple factors manuscript. Dr Moltu conceptualized and designed the study, co-
may explain the relatively low LCPUFA blood concentrations in the ordinated and supervised data collection, and critically reviewed
ARA:DHA group, such as adherence of the fatty acid supplement to and revised the manuscript. All authors approved the final manu-
the feeding tube, impaired intestinal absorption related to the use script as submitted and agreed to be accountable for all aspects of
of a non-emulsified supplementation, the unstructured timing of the work.
blood sampling in relation to fatty acid administration, or the high
number of blood transfusions. An antagonistic regulation of ARA Conflicts of interest
and DHA, in which DHA supplementation alone suppresses ARA
concentrations, seems to be a factor of importance [40]. Makrides The authors have no conflicts of interest to disclose.
27

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en enero 18, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
K. Wendel, M.F. Aas, G. Gunnarsdottir et al. Clinical Nutrition 42 (2023) 22e28

Acknowledgement [20] Lødrup Carlsen KC, Magnus P, Carlsen KH. Lung function by tidal breathing in
awake healthy newborn infants. Eur Respir J 1994;7(9):1660e8.
[21] Schmidt B, Asztalos EV, Roberts RS, Robertson CM, Sauve RS, Whitfield MF.
We would like to thank the Data Monitoring Committee mem- Impact of bronchopulmonary dysplasia, brain injury, and severe retinopathy
bers: Inge Christoffer Olsen, Ketil Stordal and Trond Markestad; Dr on the outcome of extremely low-birth-weight infants at 18 months: results
Helene Caroline Dale Osterholt for conducting Doppler echocardi- from the trial of indomethacin prophylaxis in preterms. JAMA 2003;289(9):
1124e9.
ography; Cathrine Nygaard Espeland for supervising the study; the [22] Embleton ND, Moltu SJ, Lapillonne A, van den Akker CHP, Carnielli V, Fusch C,
neonatal nurses and doctors at Oslo University Hospital for their et al. Enteral nutrition in preterm infants (2022): a position paper from the
support; and, most importantly the participating families for their ESPGHAN committee on nutrition and invited experts. J Pediatr Gastroenterol
Nutr. 9900:10.1097/MPG.0000000000003642.
commitment to the study. [23] Correani A, Dell'Orto V, Nobile S, Antognoli L, Marchionni P, Giretti I, et al.
Oxygen saturation to fraction of inspired oxygen ratio in preterm infants on
Appendix A. Supplementary data routine parenteral nutrition with conventional or fish oil containing lipid
emulsions. Pediatr Pulmonol 2020;55(9):2377e82.
[24] Bernhard W, Raith M, Koch V, Maas C, Abele H, Poets CF, et al. Developmental
Supplementary data to this article can be found online at changes in polyunsaturated fetal plasma phospholipids and feto-maternal
https://doi.org/10.1016/j.clnu.2022.11.012. plasma phospholipid ratios and their association with bronchopulmonary
dysplasia. Eur J Nutr 2016;55(7):2265e74.
[25] Ryan A, Godson C. Lipoxins: regulators of resolution. Curr Opin Pharmacol
References 2010;10(2):166e72.
[26] Martin CR, Zaman MM, Gilkey C, Salguero MV, Hasturk H, Kantarci A, et al.
[1] Stoll BJ, Hansen NI, Bell EF, Walsh MC, Carlo WA, Shankaran S, et al. Trends in Resolvin D1 and lipoxin A4 improve alveolarization and normalize septal wall
care practices, morbidity, and mortality of extremely preterm neonates, 1993- thickness in a neonatal murine model of hyperoxia-induced lung injury. PLoS
2012. JAMA 2015;314(10):1039e51. One 2014;9(6):e98773.
[2] Jobe AH. Mechanisms of lung injury and bronchopulmonary dysplasia. Am J [27] Blundell PDM, Chakraborty M. Relationship between apgar scores and
Perinatol 2016;33(11):1076e8. morbidity and mortality outcomes in preterm infants: a single-centre cohort
[3] Paananen R, Husa AK, Vuolteenaho R, Herva R, Kaukola T, Hallman M. Blood study. Neonatology 2020;117(6):742e9.
cytokines during the perinatal period in very preterm infants: relationship of [28] Lapcharoensap W, Gage SC, Kan P, Profit J, Shaw GM, Gould JB, et al. Hospital
inflammatory response and bronchopulmonary dysplasia. J Pediatr variation and risk factors for bronchopulmonary dysplasia in a population-
2009;154(1):39e43. e3. based cohort. JAMA Pediatr 2015;169(2):e143676.
[4] Savani RC. Modulators of inflammation in bronchopulmonary dysplasia. [29] Phalen AG, Kirkby S, Dysart K. The 5-minute Apgar score: survival and short-
Semin Perinatol 2018;42(7):459e70. term outcomes in extremely low-birth-weight infants. J Perinat Neonatal Nurs
[5] Leuti A, Maccarrone M, Chiurchiù V. Proresolving lipid mediators: endogenous 2012;26(2):166e71.
modulators of oxidative stress. Oxid Med Cell Longev 2019;2019:8107265. [30] Parry G, Tucker J, Tarnow-Mordi W. Crib II: an update of the clinical risk index
[6] Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. for babies score. Lancet 2003;361(9371):1789e91.
Nature 2014;510(7503):92e101. [31] Hjalmarson O, Sandberg KL. Lung function at term reflects severity of bron-
n RI, Sa
[7] Jae nchez-García S, Ferna ndez-Velasco M, Bosca L, Prieto P. Resolution- chopulmonary dysplasia. J Pediatr 2005;146(1):86e90.
based therapies: the potential of lipoxins to treat human diseases. Front [32] Bentsen MH, Markestad T, Oymar K, Halvorsen T. Lung function at term in
Immunol 2021;12:658840. extremely preterm-born infants: a regional prospective cohort study. BMJ
[8] Calder PC. Omega-3 fatty acids and inflammatory processes. Nutrients Open 2017;7(10):e016868.
2010;2(3):355e74. [33] Hellstro€m A, Pivodic A, Gra €nse L, Lundgren P, Sjo€ bom U, Nilsson AK, et al.
[9] Lapillonne A, Jensen CL. Reevaluation of the DHA requirement for the pre- Association of docosahexaenoic acid and arachidonic acid serum levels with
mature infant. Prostaglandins Leukot Essent Fatty Acids 2009;81(2e3): retinopathy of prematurity in preterm infants. JAMA Netw Open 2021;4(10):
143e50. e2128771.
[10] Martin CR, Dasilva DA, Cluette-Brown JE, Dimonda C, Hamill A, Bhutta AQ, [34] Pawlik D, Lauterbach R, Walczak M, Hurkala J, Sherman MP. Fish-oil fat
et al. Decreased postnatal docosahexaenoic and arachidonic acid blood levels emulsion supplementation reduces the risk of retinopathy in very low birth
in premature infants are associated with neonatal morbidities. J Pediatr weight infants: a prospective, randomized study. JPEN - J Parenter Enter Nutr
2011;159(5):743e9. e1-2. 2014;38(6):711e6.
[11] Boehm G, Borte M, Bo €hles HJ, Müller H, Kohn G, Moro G. Docosahexaenoic and [35] Tu CF, Lee CH, Chen HN, Tsao LY, Chen JY, Hsiao CC. Effects of fish oil-
arachidonic acid content of serum and red blood cell membrane phospho- containing lipid emulsions on retinopathy of prematurity in very low birth
lipids of preterm infants fed breast milk, standard formula or formula sup- weight infants. Pediatr Neonatol 2020;61(2):224e30.
plemented with n-3 and n-6 long-chain polyunsaturated fatty acids. Eur J [36] Beken S, Dilli D, Fettah ND, Kabatas EU, Zenciroglu A, Okumus N. The influence
Pediatr 1996;155(5):410e6. of fish-oil lipid emulsions on retinopathy of prematurity in very low birth
[12] De Rooy L, Hamdallah H, Dyall SC. Extremely preterm infants receiving weight infants: a randomized controlled trial. Early Hum Dev 2014;90(1):
standard care receive very low levels of arachidonic and docosahexaenoic 27e31.
acids. Clin Nutr 2017;36(6):1593e600. [37] Bernabe-Garcia M, Villegas-Silva R, Villavicencio-Torres A, Calder PC, Rodri-
[13] Manley BJ, Makrides M, Collins CT, McPhee AJ, Gibson RA, Ryan P, et al. High- guez-Cruz M, Maldonado-Hernandez J, et al. Enteral docosahexaenoic acid
dose docosahexaenoic acid supplementation of preterm infants: respiratory and retinopathy of prematurity: a randomized clinical trial. JPEN - J Parenter
and allergy outcomes. Pediatrics 2011;128(1):e71e7. Enter Nutr 2019;43(7):874e82.
[14] Marc I, Piedboeuf B, Lacaze-Masmonteil T, Fraser W, M^ asse B, Mohamed I, [38] Hellstro€m A, Nilsson AK, Wackernagel D, Pivodic A, Vanpee M, Sjo € bom U, et al.
et al. Effect of maternal docosahexaenoic acid supplementation on broncho- Effect of enteral lipid supplement on severe retinopathy of prematurity: a
pulmonary dysplasia-free survival in breastfed preterm infants: a randomized randomized clinical trial. JAMA Pediatr 2021;175(4):359e67.
clinical trial. JAMA 2020;324(2):157e67. [39] Frost BL, Patel AL, Robinson DT, Berseth CL, Cooper T, Caplan M. Randomized
[15] Collins CT, Makrides M, McPhee AJ, Sullivan TR, Davis PG, Thio M, et al. Do- controlled trial of early docosahexaenoic acid and arachidonic acid enteral
cosahexaenoic acid and bronchopulmonary dysplasia in preterm infants. supplementation in very low birth weight infants. J Pediatr 2021;232. 23-
N Engl J Med 2017;376(13):1245e55. 30.e1.
[16] Colombo J, Jill Shaddy D, Kerling EH, Gustafson KM, Carlson SE. Docosahex- [40] Hsieh AT, Anthony JC, Diersen-Schade DA, Rumsey SC, Lawrence P, Li C, et al.
aenoic acid (DHA) and arachidonic acid (ARA) balance in developmental The influence of moderate and high dietary long chain polyunsaturated fatty
outcomes. Prostaglandins Leukot Essent Fatty Acids 2017;121:52e6. acids (LCPUFA) on baboon neonate tissue fatty acids. Pediatr Res 2007;61(5 Pt
[17] Wendel K, Pfeiffer HCV, Fugelseth DM, Nestaas E, Domello €f M, Skålhegg BS, 1):537e45.
et al. Effects of nutrition therapy on growth, inflammation and metabolism in [41] Makrides M, Neumann MA, Simmer K, Gibson RA. Erythrocyte fatty acids of
immature infants: a study protocol of a double-blind randomized controlled term infants fed either breast milk, standard formula, or formula supple-
trial (ImNuT). BMC Pediatr 2021;21(1):19. mented with long-chain polyunsaturates. Lipids 1995;30(10):941e8.
[18] Mihatsch WA, Braegger C, Bronsky J, Cai W, Campoy C, Carnielli V, et al. [42] Diau GY, Hsieh AT, Sarkadi-Nagy EA, Wijendran V, Nathanielsz PW, Brenna JT.
ESPGHAN/ESPEN/ESPR/CSPEN guidelines on pediatric parenteral nutrition. The influence of long chain polyunsaturate supplementation on docosahex-
Clin Nutr 2018;37(6 Pt B):2303e5. aenoic acid and arachidonic acid in baboon neonate central nervous system.
[19] Higgins RD, Jobe AH, Koso-Thomas M, Bancalari E, Viscardi RM, Hartert TV, BMC Med 2005;3:11.
et al. Bronchopulmonary dysplasia: executive summary of a workshop. [43] Piomelli D. Arachidonic acid in cell signaling. Curr Opin Cell Biol 1993;5(2):
J Pediatr 2018;197:300e8. 274e80.

28

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en enero 18, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.

You might also like