oulu 2018 d 1480 university of oulu p.o. box 8000 fi...

90
UNIVERSITATIS OULUENSIS MEDICA ACTA D D 1480 ACTA Pia Härkin OULU 2018 D 1480 Pia Härkin CLOSURE OF PATENT DUCTUS ARTERIOSUS IN VERY PRETERM INFANTS POTENTIAL ROLE OF PARACETAMOL AND CONSEQUENCES OF CURRENT TREATMENTS UNIVERSITY OF OULU GRADUATE SCHOOL; UNIVERSITY OF OULU, FACULTY OF MEDICINE: MEDICAL RESEARCH CENTER OULU; OULU UNIVERSITY HOSPITAL

Upload: others

Post on 27-Feb-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

UNIVERSITY OF OULU P .O. Box 8000 F I -90014 UNIVERSITY OF OULU FINLAND

A C T A U N I V E R S I T A T I S O U L U E N S I S

University Lecturer Tuomo Glumoff

University Lecturer Santeri Palviainen

Postdoctoral research fellow Sanna Taskila

Professor Olli Vuolteenaho

University Lecturer Veli-Matti Ulvinen

Planning Director Pertti Tikkanen

Professor Jari Juga

University Lecturer Anu Soikkeli

Professor Olli Vuolteenaho

Publications Editor Kirsti Nurkkala

ISBN 978-952-62-2024-6 (Paperback)ISBN 978-952-62-2025-3 (PDF)ISSN 0355-3221 (Print)ISSN 1796-2234 (Online)

U N I V E R S I TAT I S O U L U E N S I S

MEDICA

ACTAD

D 1480

AC

TAP

ia Härkin

OULU 2018

D 1480

Pia Härkin

CLOSURE OF PATENT DUCTUS ARTERIOSUS IN VERY PRETERM INFANTSPOTENTIAL ROLE OF PARACETAMOL AND CONSEQUENCES OF CURRENT TREATMENTS

UNIVERSITY OF OULU GRADUATE SCHOOL;UNIVERSITY OF OULU,FACULTY OF MEDICINE:MEDICAL RESEARCH CENTER OULU;OULU UNIVERSITY HOSPITAL

Page 2: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus
Page 3: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

ACTA UNIVERS ITAT I S OULUENS I SD M e d i c a 1 4 8 0

PIA HÄRKIN

CLOSURE OF PATENT DUCTUS ARTERIOSUS IN VERY PRETERM INFANTSPotential role of paracetamol and consequences of current treatments

Academic dissertation to be presented with the assent ofthe Doctoral Training Committee of Health andBiosciences of the University of Oulu for public defence inAuditorium 12 of Oulu University Hospital, on 16November 2018, at 12 noon

UNIVERSITY OF OULU, OULU 2018

Page 4: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

Copyright © 2018Acta Univ. Oul. D 1480, 2018

Supervised byProfessor Mikko HallmanDocent Timo Saarela

Reviewed byDocent Tiina OjalaDoctor Viena Tommiska

ISBN 978-952-62-2024-6 (Paperback)ISBN 978-952-62-2025-3 (PDF)

ISSN 0355-3221 (Printed)ISSN 1796-2234 (Online)

Cover DesignRaimo Ahonen

JUVENES PRINTTAMPERE 2018

OpponentDocent Hanna Soukka

Page 5: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

Härkin, Pia, Closure of patent ductus arteriosus in very preterm infants. Potentialrole of paracetamol and consequences of current treatmentsUniversity of Oulu Graduate School; University of Oulu, Faculty of Medicine, MedicalResearch Center Oulu; Oulu University HospitalActa Univ. Oul. D 1480, 2018University of Oulu, P.O. Box 8000, FI-90014 University of Oulu, Finland

Abstract

The ductus arteriosus connects the pulmonary artery and the descending aorta in the foetus. Innormal neonatal transition, the ductus closes soon after birth. If the duct remains significantly openafter birth, it may complicate the recovery of a very preterm infant. Present treatments of patentductus arteriosus (PDA) are either medical (ibuprofen or indomethacin) or surgical (ligation).However, these treatments can have serious side effects, especially in the most immature infants.This doctoral thesis studied the potential role of intravenous paracetamol for PDA treatment invery preterm infants born before 32 weeks of gestation. Consequences of the PDA treatments inan epidemiological birth cohort were also studied. In retrospective Study I stated that treatmentsof PDA decreased after the introduction of IV paracetamol for early pain management in preterminfants. Study II showed in a randomised clinical trial for the first time that paracetamol has abiological effect on the ductus arteriosus in preterm infants soon after birth. The ductus closedsignificantly earlier in the paracetamol group than in the placebo group. The epidemiologicalcohort Study III showed evidence that both medical and surgical treatment of PDA associated withsevere bronchopulmonary dysplasia in infants born very preterm. Additionally, surgical PDAligation was associated with increased risk of necrotising enterocolitis and intraventricularhaemorrhage. Study IV showed that treatment of PDA was not associated with increasedmortality, even in the most immature preterm infants born before 28 weeks of gestation.

Keywords: cohort study, morbidity, paracetamol, patent ductus arteriosus, PDAepidemiology, PDA treatment, very low gestational age infant

Page 6: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus
Page 7: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

Härkin, Pia, Hyvin pienen keskosen avoimen valtimotiehyen sulku. Parasetamoli-hoidon merkitys ja nykyhoitojen sivuvaikutuksetOulun yliopiston tutkijakoulu; Oulun yliopisto, Lääketieteellinen tiedekunta; Medical ResearchCenter Oulu; Oulun yliopistollinen sairaalaActa Univ. Oul. D 1480, 2018Oulun yliopisto, PL 8000, 90014 Oulun yliopisto

Tiivistelmä

Valtimotiehyt on sikiöaikana avoimena oleva suoni, joka yhdistää keuhkovaltimon laskevaanaorttaan ja ohjaa vähähappisen veren istukkaan. Yhdessä soikean aukon kanssa suoni takaa siki-ön verenkierron normaalin toiminnan ennen keuhkojen avautumista. Mikäli valtimotiehyt jääsyntymän jälkeen pitkittyneesti auki, muuttaa se keskosen verenkiertoa siten, että osa aortanverenkiertoa ohjautuu keuhkoverenkiertoon vaikeuttaen pienen keskosen toipumista. Nykyhoi-toina käytetään joko lääkkeellistä (ibuprofeeni tai indometasiini) tai kirurgista sulkua. Lääkkeel-linen hoito ei ole kovin tehokas kaikista epäkypsimmillä keskosilla ja hoitoihin liittyy vakavia-kin sivuvaikutuksia.

Väitöskirjassa tutkittiin parasetamolilääkityksen vaikutusta hyvin pienen keskosen avoimenvaltimotiehyen sulkeutumiseen. Epidemiologisessa osiossa tutkittiin nykyhoitojen sivuvaikutuk-sia hyvin pienillä keskosilla. Osatyössä I todettiin, että avoimen valtimotiehyen hoidon tarveväheni merkittävästi sen jälkeen kun parasetamoli oli otettu käyttöön kivun hoidossa vastasynty-neiden teholla. Osatyö II oli satunnaistettu ja sokkoutettu hoitotutkimus, jossa todettiin alkupe-räishavaintona, että parasetamolilla on biologinen vaikutus keskosen avoimeen valtimotiehyeen.Parasetamolia saaneilla keskosilla valtimotiehyt sulkeutui aikaisemmin kuin verrokeilla. Hoidol-la ei todettu merkittäviä sivuvaikutuksia. Osatöissä III ja IV tutkittiin kaikkien vuosina2005−2013 Suomessa syntyneiden hyvin pienten keskosten avoimen valtimotiehyen hoitoja.Lääkehoidolla (ibuprofeeni ja indometasiini) ja kirurgisella hoidolla todettiin olevan yhteys kes-kosen kroonisen keuhkotaudin (BPD) vaikeimpaan muotoon. Kirurgisella hoidolla oli yhteyskeskosen vaikeaan suolitulehdukseen ja vaikeaan aivoverenvuotoon. Kuolleisuuden riskin eikuitenkaan todettu lisääntyneen valtimotiehyen hoitoihin liittyen.

Asiasanat: avoimen valtimotiehyen hoito, avoin valtimotiehyt, hyvin pieni keskonen,kohorttitutkimus, kuolleisuus, parasetamoli

Page 8: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus
Page 9: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

To my family

Page 10: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

8

Page 11: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

9

Acknowledgements This work was carried out in the Department of Children and Adolescents, Oulu

University Hospital and University of Oulu, during 2013–2018.

I wish to express my deepest gratitude to my supervisors, Professor Mikko

Hallman and Docent Timo Saarela. Mikko, your knowledge of research is very

fascinating and I truly admire your long and successful international career as a

scientist. Timo’s sharp clinical skills have delighted me during these years at

NICU. No matter if it is day or night, you have always given me an answer to any

clinical problem. Thank you Timo, you have taught me so much about

neonatology. I would like to offer my special thanks to Docent Päivi Tapanainen,

Head of the Division of Paediatrics and Gynaecology, for nice discussions and the

benevolence towards me during these years. I wish to thank Professor Mika

Rämet for providing me good facilities for the clinical research. I am deeply

grateful for my first teacher in the field of neonatology, Docent Marja-Leena

Pokela. I also want to thank Mikko Remes for your essential guidance and

teaching which I received as a young doctor. I am grateful for Professor Matti

Uhari for introducing me to the world of EBM. These repeated lessons finally

reached the target.

I sincerely thank the official referees of the thesis, Docent Tiina Ojala and

Viena Tommiska, for their wise and constructive comments.

Special thanks go to Tytti Pokka for her eternal patience and guidance with

the statistics. Tytti, without you I would still sit in front of my computer staring at

the wonderful SPSS software and the everlasting epidemiological files. My

closest co-authors Outi Aikio and Docent Riitta Marttila earn my deepest

gratitude. I thank all the members of the follow-up group, Docent Terhi

Tapiainen, Docent Tuula Kaukola, and Docent Matti Nuutinen for your great

advices on my research. Tuula, my roommate at work, your kindness and wise

advice served with chocolate have been very important and sometimes even

lifesaving. Special thanks go to Docent Marita Valkama for teaching me both

neonatological skills and the very essential theoretical part. I wish to thank Antti

Härmä for your collegial attitude towards me during these years. All the rest of

the colleagues in the Department of Paediatrics at Oulu University Hospital which

I have worked with during these years, thank you. Special thanks go to Sami

Turunen, Merja Kallio and Docent Marja Ojaniemi.

Page 12: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

10

I wish to thank you the unbelievably nice, clever, funny and professional

nurses in the ward of our NICU. Riitta Vikeväinen, your work as study nurse in

the Preparas study was very important, thank you so much.

Kaikukuoro members, our choir has brought me so many joyful moments

during these years. I am so happy to have the music and this warm-hearted group

in my life, you ALL are important. My friends since childhood Tiina, Marja and

Kaisa, without you all I would not be me, in a positive way. My colleagues from

the Royal Prosecco Club, Mervi Linna and Johanna Uunila, have the great

privilege to be mentioned as royal ladies, you are all that and so much more,

thank you for everything. You are awesome ladies and fantastic human beings. I

owe my deepest gratitude to my colleagues and friends Docent Outi Peltoniemi,

and Katariina Mankinen. You have always been there, I am deeply grateful for

you both of our close friendship. My precious colleague and dear friend Efthymia

Protonotariou we have had so warm and nice relationship during these years

despite of the distance. Thank you Efi for your hospitality, kindness and the

fantastic philosophical discussions we have had over the years.

This life would be empty, lonely and senseless without my family. My

parents, my mother and my late father always believed in me and supported me,

thank you so much. You offered me a great childhood with so many activities,

which gave me a wide perspective to life itself. Mother, you were always there

when I needed you, thank you. I want to thank my brother Ismo and my sister

Päivikki for everything. The whole sister’s family, you all are so awesome and

fantastic people! My beloved children Paavo and Tatu, you have shown me the

true meaning of life and love, thank you both of my precious ones. Mother loves

you so much. And finally, thank you dear Mika so much for everything. Our first

years with children and the sudden losses of our close ones were not the easiest

ones, but finally we survived after all those heavy years. This is a true merit!

This work was supported financially by the Alma and K.A. Foundation, the

University of Oulu Graduate School, the Foundation of Paediatric Research and

the Medical Research Center, Oulu.

Oulu, September 2018 Pia Härkin

Page 13: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

11

Abbreviations AA arachidonic acid

APAP acetyl-para-aminophenol

BW birth weight

BNP brain natriuretic peptide

BPD bronchopulmonary dysplasia

CI confidence intervals

CLD chronic lung disease

COX cyclooxygenase

DA ductus arteriosus

ECHO echocardiography

ELGA extremely low gestational age (< 28 weeks)

GA gestational age

hsPDA hemodynamically significant patent ductus arteriosus

IV intravenous

IVH intraventricular haemorrhage

LA/Ao left-atrium-to-aorta ratio

LPA left pulmonary artery

MCA middle cerebral artery

MPA main pulmonary artery

NAPQI N-acetyl-p-benzoquinone-imine

NDI neurodevelopmental impairment

NEC necrotising enterocolitis

NSAID non-steroidal anti-inflammatory drug

NNT number needed to treat

NT-proBNP amino-terminal pro-B-type natriuretic peptide

OR odds ratio

PAH pulmonary arterial hypertension

PDA patent ductus arteriosus

PG prostaglandin

PGE2 prostaglandin E2

PLCS post-ligation cardiac syndrome

PROM premature rupture of the foetal membranes

PVL periventricular leukomalacia

RCT randomised controlled trial

RDS respiratory distress syndrome

Page 14: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

12

ROP retinopathy of prematurity

RR relative risk

SD standard deviation

SGA small for gestational age

VLGA very low gestational age (< 32 weeks)

Page 15: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

13

List of original articles This thesis is based on the following publications, which are referred throughout

the text by their Roman numerals:

I Aikio O., Härkin P., Saarela T., & Hallman M. (2014). Early paracetamol treatment associated with lowered risk of persistent ductus arteriosus in very preterm infants. J Matern Fetal Neonatal Med, 27(12), 1252-6.

II Härkin P., Härmä A., Aikio O., Valkama M., Leskinen M., Saarela T. & Hallman M. (2016). Paracetamol Accelerates Closure of the Ductus Arteriosus after Premature Birth: A Randomized Trial. J Pediatr. 177, 72-77.e2.

III Härkin P., Marttila R., Pokka T., Saarela T., & Hallman M. (2017) Morbidities associated with patent ductus arteriosus in preterm infants. Nationwide cohort study. J Matern Fetal Neonatal Med. 11, 1-8. doi: 10.1080/14767058.2017.1347921.

IV Härkin P., Marttila R., Pokka T., Saarela T., & Hallman M. (2018). National survival analysis of infants born extremely preterm. Manuscript.

Page 16: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

14

Page 17: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

15

Table of contents Abstract

Tiivistelmä

Acknowledgements 9 Abbreviations 11 List of original articles 13 Table of contents 15 1 Introduction 17 2 Review of the literature 19

2.1 Physiology and pathophysiology of the ductus arteriosus ...................... 19 2.2 The incidence of patent ductus arteriosus (PDA) .................................... 20 2.3 Diagnosis and definition of hemodynamically significant PDA ............. 21

2.3.1 Echocardiography (ECHO) .......................................................... 21 2.3.2 Clinical diagnosis of hsPDA in very preterm infants ................... 22 2.3.3 Postnatal age of ductal ECHO screening for treatment ................ 22 2.3.4 Biochemical markers .................................................................... 23

2.4 Treatment of PDA ................................................................................... 23 2.4.1 Treatment challenges .................................................................... 23 2.4.2 Conservative management of PDA .............................................. 24 2.4.3 Medical treatment of PDA ............................................................ 25 2.4.4 Surgical ligation of PDA .............................................................. 28 2.4.5 Other options for the treatment ..................................................... 29

2.5 Postnatal age for PDA treatment ............................................................. 30 2.5.1 Prophylactic and early treatment .................................................. 30 2.5.2 Late treatment (symptomatic therapy) .......................................... 31 2.5.3 Optimal postnatal age for treatment ............................................. 31

2.6 Paracetamol (acetaminophen) ................................................................. 32 2.6.1 Paracetamol for pain in preterm infants ....................................... 32 2.6.2 Paracetamol for patent ductus arteriosus ...................................... 35 2.6.3 Side effects of paracetamol ........................................................... 38

2.7 Association of the treatments of PDA with neonatal morbidities ........... 39 2.7.1 Associations with bronchopulmonary dysplasia (BPD) ............... 40 2.7.2 Associations with other neonatal outcomes and mortality ........... 41

3 Aims of the research 43 4 Patients and methods 45

4.1.1 Paracetamol for patent ductus arteriosus (I and II) ....................... 45

Page 18: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

16

4.1.2 Morbidities and mortality associated with PDA treatments

(III) ............................................................................................... 49 4.1.3 National survival analysis of infants born extremely

preterm (IV). ................................................................................. 52 5 Results 53

5.1 Paracetamol for patent ductus arteriosus ................................................. 53 5.2 Morbidities and mortality associated with PDA treatments (III) ............ 57 5.3 Mortality of infants born extremely preterm (IV) ................................... 61

6 Discussion 65 7 Conclusions 69 References 71 Original publications 85

Page 19: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

17

1 Introduction The rate of preterm birth varies globally, occurring in 5–18% of all births and

contributing significantly to infant mortality and morbidity (Blencowe et al.,

2012). Prematurity is the largest cause of neonatal death worldwide (Kong et al.,

2016; Liu et al., 2016). The risk for mortality and morbidity is highest in infants

born extremely preterm, under i.e. less than 28 weeks of gestation (EXPRESS

Group et al., 2009; Serenius et al., 2014). Mortality and morbidity rates of very

preterm infants differ between regions and countries (Boghossian, Geraci,

Edwards, & Horbar, 2018; Bonet et al., 2017; Edstedt Bonamy et al., 2018;

Serenius et al., 2014).

Level of immaturity is the main contributing factor to later neonatal morbidity,

such as retinopathy of prematurity (ROP), necrotising enterocolitis (NEC),

intraventricular haemorrhage (IVH), and bronchopulmonary dysplasia (BPD).

However, the importance and the independent role of the patent ductus arteriosus

(PDA) in the later management and survival of preterm infants are controversial

and unclear. Therapies used for the closure of PDA are either medical (ibuprofen

or indomethacin) or surgical, but these therapies have potential side effects for

premature infants (R. I. Clyman & Chorne, 2007; Janz-Robinson et al., 2015;

Madan, Kendrick, Hagadorn, Frantz, & National Institute of Child Health and

Human Development Neonatal Research Network, 2009). There is no consensus

regarding either the criteria of hemodynamically significant patent ductus

arteriosus (hsPDA) or the optimal postnatal age of treatment (Jain & Shah, 2015;

Skelton, Evans, & Smythe, 1994; Zonnenberg & de Waal, 2012).

Previous case reports and observational studies have shown that paracetamol

may be an alternative to the closure of PDA (Allegaert, 2013; Aminoshariae &

Khan, 2015; Hammerman et al., 2011; Oncel et al., 2013). Several recent

randomised controlled trial (RCT) studies also show that paracetamol may be as

effective as ibuprofen and indomethacin for closing PDA of preterm infants, but

with fewer side effects. However, evidence of the indications, dosage,

effectiveness, and safety −including the long-term effects of paracetamol− are still

incomplete or lacking.

Page 20: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

18

Page 21: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

19

2 Review of the literature

2.1 Physiology and pathophysiology of the ductus arteriosus

Patency of the ductus arteriosus (DA) is essential in the foetal period, as it

connects the main pulmonary artery (MPA) to the descending aorta (Ao) to

guarantee normal foetal circulation and development of the foetus. The DA

diverts ventricular output away from the lungs and towards the placenta (Hamrick

& Hansmann, 2010). Closure of the duct during the foetal period may lead to

foetal demise. In normal situations, the duct acts like a physiological shunt and

closes functionally by 72 hours of age in 90% of term infants (Gentile et al., 1981;

Skinner, Skinner, Alverson, & Hunter, 2000). Normally, the shunt pattern changes

left to right before complete closure (Skelton et al., 1994). In normal functional

closure of PDA, the wall of the vessel thickens, endothelial cells activate, and

smooth muscle cells contract. Vasodilatory agents drop (the main vasodilator is

PGE2) and contracting agents (e.g, oxygen level) rise, resulting in the vessel

contracting functionally (Hamrick & Hansmann, 2010). In normal situations, after

functional closure, the anatomical (final) closure takes place within three weeks

of age. In most preterm infants, the functional closure of PDA is immature and

delayed.

If the DA remains open for a prolonged period after birth, it can change from

a physiological to a pathophysiological shunt, or PDA. Prolonged patency of the

duct can be caused by structural, functional, immature, and altered physiological

conditions (Bokenkamp, DeRuiter, van Munsteren, & Gittenberger-de Groot,

2010). Because there is normal individual variation of neonatal cardiovascular

physiology and circulation changes during the transition period, the hemodynamic

diagnostics during the early postnatal time is challenging (Vrancken, van Heijst,

& de Boode, 2018). After normal postnatal decrease of high pulmonary pressure,

the shunt direction soon changes from right to left (foetal circulation) and

becomes left to right (from the aorta to the pulmonary artery) also in immature

infants. Normally, the shunt is diverted completely or predominantly in 95% of

preterm infants by 5 hours after birth in infants born under 30 gestation weeks

(Kluckow & Evans, 2000b). If PDA stays hemodynamically significantly open

(hsPDA), it may lead to overloading in the right side (pulmonary overcirculation)

and systemic hypoperfusion in the left side (low systemic blood flow) (R. I.

Page 22: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

20

Clyman, 2012). This change in circulation may induce symptoms in the infant

condition.

The right-side overloading via the ductal shunt may lead to pulmonary

problems, including prolonged need of mechanical ventilation, surfactant

inhibition, pulmonary oedema or haemorrhage and later, the risk of BPD and

death (Kluckow & Evans, 2000a). On the left side, systemic hypoperfusion may

lead to end-organ hypoperfusion and ischemia as a result of reduced renal and

mesenteric flow, reduced middle cerebral artery blood flow velocity, and,

thereafter, possible risk for NEC, IVH and periventricular leukomalacia (PVL)

(Evans & Iyer, 1994; Evans & Kluckow, 1996; Evans, 2015; Pladys et al., 2001).

Large PDA can sometimes lead to cardiac failure, but this, fortunately, is rare.

Association of PDA with later morbidities is not clear however, and does have

great individual variability. Comorbidities depend on the size of PDA, the degree

of immaturity, and the capacity of the compensatory mechanisms of the infant’s

hemodynamic system. For example, when cerebral autoregulation is disturbed,

the risk for IVH with PDA rises (Perlman, Hill, & Volpe, 1981; Van Bel, Van de

Bor, Stijnen, Baan, & Ruys, 1989). Even large PDA with low systemic blood

pressure combined with good cerebral autoregulation, does not necessarily

influence the infant’s prognosis. However, the single or independent role of PDA

and its relationship to later morbidities is not clear (Hamrick & Hansmann, 2010).

2.2 The incidence of patent ductus arteriosus (PDA)

In healthy term infants, the incidence of PDA is between 0.02% and 0.006% of

live births, with a 2:1 female-to-male ratio (Tripathi, Black, Park, & Jerrell, 2013).

The aetiology is suggested to be multifactorial, with some genetic component in

the pathogenesis (Bokenkamp et al., 2010; Tripathi et al., 2013). In preterm

infants, the occurrence of PDA is inversely related to gestational weeks. The more

premature the infant, the higher the incidence of PDA. In very low gestational age

(VLGA) infants (born < 32 weeks), more than 30% have PDA, and in those born

under 28 weeks, 55–70% are at risk for significant open ductus requiring

treatment (Reller, Rice, & McDonald, 1993). However, spontaneous closure is

frequent. By 7 days of life, about 80% of infants born between 26 and 31

gestational weeks had no or only small PDA (Van Overmeire, Van de Broek, Van

Laer, Weyler, & Vanhaesebrouck, 2001). In contrast, in the group of infants born

<25 gestational weeks, the proportion of spontaneous closure was only 12.5%,

Page 23: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

21

showing that the most immature infants are at the highest risk for PDA (Koch et

al., 2006).

2.3 Diagnosis and definition of hemodynamically significant PDA

2.3.1 Echocardiography (ECHO)

Colour Doppler heart echocardiography (ECHO) is used to measure the

hemodynamic significance and the volume of the ductal shunt (ductal size).

ECHO should be performed prior to closing the duct to reveal possible congenital

heart defect (Chu, Li, Kosinski, Hornik, & Hill, 2017) and related

contraindications for ductal closure. In case of ductal-dependent systemic or

pulmonary circulation, closing the duct can be life threatening for the infant. In

this case, prostaglandin analogues must be infused to keep the ductal vessel open.

The parameters and ECHO criteria for hsPDA are variable (Jain & Shah,

2015). There are criteria and scoring of hsPDA, but without clear consensus (A.

El-Khuffash et al., 2015; Sehgal, Paul, & Menahem, 2013; Skelton et al., 1994;

Su, Watanabe, Shimizu, & Yanagisawa, 1997). ECHO parameters commonly used

for hsPDA include PDA diameter (the internal size of the duct in two-dimensional

view > 1.5-2.0 mm or 2 mm/kg), the left-atrium-to-aortic root ratio (LA/Ao > 1.4),

internal ductal diameter > 50% wider than that of the left pulmonary artery,

velocity of the shunt < 2 m/s, and a flow pattern characterised by a large left-to-

right ductal shunt (Evans & Iyer, 1994; Skinner et al., 2000; Zonnenberg & de

Waal, 2012). In clinical practice, evaluating ductal flow and the vessel’s true

diameter is challenging because vessel dimensions vary (diameter is not static)

during the transitional period, especially in the most immature infants. There is

also anatomical and topological variance in the ductal architecture and viewing

three-dimensional structures in two-dimension ECHO requires experience and

expertise (Krichenko et al., 1989). Left-side (atrial or ventricular) dilatation

indicates that the ductus is hemodynamically significant. Large PDA for

prolonged time may lead to heart failure. Pressure gradient across PDA shows a

continuous flow profile from left to right, indicating a possible large- volume

shunt (Evans & Iyer, 1994; Skinner et al., 2000). Experienced

echocardiographists and paediatric cardiologists employ other detailed ECHO

parameters, such as pulmonary vein Doppler measurements and pulmonary

overcirculation measurements from left ventricular output overload. In

Page 24: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

22

large/moderate-volume ductal shunt systemic hypoperfusion (low systemic blood

flow), signs can be seen from absent or reversed diastolic flow from the middle

cerebral artery, the descending aorta, or/and the coeliac trunk artery (A. F. El-

Khuffash, Jain, & McNamara, 2013).

2.3.2 Clinical diagnosis of hsPDA in very preterm infants

The clinical criteria and symptoms of hsPDA in premature infants are classified as

a murmur, cardiopulmonary distress, hyperdynamic precordium, bounding pulses,

increased need for respiratory support, and wide pulse pressure. A PDA murmur

is continuous if there is a constant pressure gradient in both the systole and the

diastole as blood is forced via the duct from left to right (from the aorta into the

pulmonary artery). The aortic blood pressure must be higher than the pulmonary

pressure during diastole to cause the left to right shunt direction in the ductal flow.

McNamara’s criteria combine clinical signs and ECHO parameters (McNamara &

Sehgal, 2007). Another scoring system for PDA defines the significance of PDA

based on ECHO criteria (A. El-Khuffash et al., 2015). The challenge with these

definitions is that there is still no consensus for clinicians regarding hsPDA

criteria, which PDA should be treated, when, and how. In addition, the great

individual variation in premature infants’ hemodynamical adaptance to the PDA

shunt makes the criteria for treatment challenging for the clinician. Decisions

about therapy should be based on individual clinical signs of a large left-right

shunt with echocardiographic parameters showing hsPDA. Chest x-ray may also

show signs of increased pulmonary vasculature and notable heart dilatation in the

case of hsPDA.

2.3.3 Postnatal age of ductal ECHO screening for treatment

There are different approaches regarding the optimal postnatal age for ECHO

screening and the subsequent treatment procedures for PDA in preterm infants.

Potential approaches to PDA management are early screening, early targeted

intervention, or delayed intervention, which indicates a more conservative attitude.

Early screening (before day 3 of life) in extremely low gestational age (ELGA)

infants was associated with lower in-hospital mortality (number needed to treat

(NNT) to prevent one death = 23), but there were no differences between major

neonatal morbidities such as NEC, severe BPD, or severe cerebral lesions when

compared to unscreened infants (Roze et al., 2015). Performed on second

Page 25: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

23

postnatal day, cardiac ultrasound-targeted measures of PDA diameter and

maximum flow velocity in very preterm infants showed that a specific PDA

severity score is able to predict the later occurrence of chronic lung disease (CLD)

or death in the high-risk group (A. El-Khuffash et al., 2015). Early screening

entails a risk of overtreatment of PDA and a possible higher risk for adverse

effects associated with the treatment procedures. Early targeted intervention of

ductal closure is based mainly on shunt volume, and its benefits are the selection

of patients with an evolving shunt. The later or delayed (conservative)

intervention approach is based mainly on clinical assessment, and entails less

need for treatment but also a potential risk for later morbidity (risk for BPD) and

mortality associated with PDA itself.

2.3.4 Biochemical markers

Brain natriuretic peptide (BNP) and amino-terminal pro-B type natriuretic peptide

(NT-proBNP) are synthesised and released into the circulatory system by the

cardiac ventricular myocytes in response to pressure overload, volume expansion

and increased myocardial wall stress. Because the hemodynamic efficient of PDA

is occasionally difficult to comprehend with a sole ECHO, a simple blood test

would provide an attractive additional diagnostic tool for clinicians. Unfortunately,

studies have shown that the diagnostic accuracy of BNP and NT-Pro-BNP for

hsPDA are not valid for clinical decisions in the management of hsPDA of

premature infants (M. Kulkarni et al., 2015).

2.4 Treatment of PDA

2.4.1 Treatment challenges

Medical treatment with either indomethacin or ibuprofen and surgical closure

(PDA ligation) is the mainstay in the management of PDA. Of late, conservative

management has become a more popular choice of treatment. There is no

consensus of treatment (Evans, 2015), which reflects the holistic problem of

reconciling PDA in preterm infants with the continuing dilemma of diagnosing

and defining hsPDA. The Effective Perinatal Intensive Care in Europe (EPICE)

study (a population-based cohort study in 19 regions in 11 European countries

during 2011-2012 of all births born before 32 gestational weeks) noticed that

Page 26: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

24

treatment policies of PDA treatments were very heterogenic. However, the

variation in PDA treatment did not explain differences in patient risk factors such

as mortality and severe BPD (Edstedt Bonamy et al., 2017). The independent role

of PDA and its association with later morbidities is not clear, and a more uniform

strategy is needed. The treatment of PDA is challenging as spontaneous closure is

frequent, and the solid evidence of treatment benefit is lacking. PDA treatment

trials (randomised controlled studies of PDA treatment) have not shown evidence

of a reduction in PDA-associated morbidities such as severe IVH, NEC, severe

BPD, death, and worsened neurodevelopmental outcome (Benitz, 2010; Benitz,

2012; R. I. Clyman & Chorne, 2007). The treatment of PDA and, especially the

early active closure of PDA and its association with later neonatal morbidity

remains inconclusive (A. El-Khuffash et al., 2015; Gudmundsdottir et al., 2015;

Schena et al., 2015; Slaughter, Reagan, Bapat, Newman, & Klebanoff, 2016;

Weisz, More, McNamara, & Shah, 2014). The EPICE study showed that the

proportion of PDA treatments varied from 10% to 39% (p < 0.001) between

(European) regions, indicating different management approaches (Edstedt

Bonamy et al., 2017).

2.4.2 Conservative management of PDA

The conservative treatment option means watchful waiting for spontaneous

closure of PDA. Recently, the trend of conservative management has become

more accepted approach (Benitz & Committee on Fetus and Newborn, American

Academy of Pediatrics, 2016). It has been shown that routine treatment to close

PDA, either medically or surgically, in the first two weeks after birth does not

improve long-term outcomes (level of evidence: 1A) (Benitz & Committee on

Fetus and Newborn, American Academy of Pediatrics, 2016). Neither clinical

trials nor meta-analyses have shown that closing the duct improves long-term

outcomes. Routine treatment, especially when conducted early on, has no effect

on morbidities like BPD, NEC, and mortality (Benitz, 2010). Prophylactic

indomethacin (given by 12 hours of life) seems to reduce the rate of severe IVH

and early severe pulmonary haemorrhage, but it did not improve long-term

neurodevelopmental or respiratory outcomes (Schmidt et al., 2001). Jhaveri et al.

(2010) reported a significantly lower rate of NEC in ELGA infants without

treatment of PDA (conservative management) when compared to infants treated

with early ligation after an attempt of indomethacin therapy (Jhaveri, Moon-

Grady, & Clyman, 2010). In a recent meta-analysis of different treatment options

Page 27: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

25

for hsPDA (ibuprofen, indomethacin, paracetamol or placebo), placebo or no

treatment was not associated with an increased likelihood of mortality and

morbidity such as NEC or IVH (Mitra et al., 2018). On the contrary, a multicentre

cohort study of the most preterm infants (born < 1000 g, included infants n=494)

showed that a conservative approach towards PDA was associated with higher

mortality, whereas a surgical approach was associated with an increased

occurrence of BPD at 36 weeks and ROP. However, medical and conservative

treatments were protective for the outcome of death/BPD at 36 weeks (Sadeck et

al., 2014).

High fluid intake (> 170 mL/kg/day) during the first days of life in very

premature infants has been associated with an increased risk of PDA (Stephens et

al., 2008). Fluid restriction has been shown to predispose closure of the ductus,

thus constituting an important part of conservative PDA management. Optimal

fluid restriction in VLGA infants improves pulmonary function by decreasing the

overloading of pulmonary circulation (De Buyst, Rakza, Pennaforte, Johansson,

& Storme, 2012). In a systematic review, restricted fluid intake is associated with

a reduction in PDA (E. F. Bell & Acarregui, 2008). When the fluid volume is

restricted, adequate energy intake must be evaluated to ensure sufficient calorie

intake and avoid harming the premature infant’s optimal outcome. In practice, this

means adding energy supplies such as carbohydrates, protein, and fat to the diet to

prevent growth restriction.

The use of diuretics is not recommended for the treatment of PDA because

there is no evidence of benefits to PDA closure. Furthermore, side effects of these

drugs, especially to premature infants in their early days, could present more

drawbacks than advantages (Brion & Campbell, 1999; Ketkeaw, Thaithumyanon,

& Punnahitananda, 2004; B. S. Lee et al., 2010).

2.4.3 Medical treatment of PDA

Medical treatments for PDA closure in preterm infants include the

cyclooxygenase (COX) inhibitors ibuprofen and indomethacin. These drugs are

non-steroidal anti-inflammatory drugs (NSAIDs) that contract the duct by

inhibiting the formation of vasodilators such as prostaglandins (PGs) (Figure 1).

Inhibition occurs when the drugs influence the cyclooxygenase enzyme and block

the formation of PGs (mainly PGE2) from arachidonic acid (AA) (R. I. Clyman,

Saha, Jobe, & Oh, 2007; Ohlsson, Walia, & Shah, 2015; Sadeck et al., 2014).

Treatment success is about the same (70−80%) for both ibuprofen and

Page 28: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

26

indomethacin (Gournay et al., 2004). ELGA infants, in whom the occurrence of

hsPDA is highest, present the most challenging and problematic group for

medical treatment. The efficiency of the drug is generally worse in ELGA infants

than in more mature infants. Furthermore, due to immaturity, the frequencies of

other comorbidities, and the risks of side effects due to treatments are higher in

these high-risk infants. Another dilemma is that a meta-analysis of more than 50

RCT studies found no long-term benefits regarding NSAID treatments in preterm

infants (Benitz, 2010; Benitz, 2012).

Indomethacin

Indomethacin is an NSAID that inhibits COX enzymes and prevents the

formation of PGs from AA (Figure 1). It was first described in 1976 for the

closure of PDA in premature infants (Friedman, Hirschklau, Printz, Pitlick, &

Kirkpatrick, 1976; Heymann, Rudolph, & Silverman, 1976). Indomethacin is the

most widely used COX-inhibitor for PDA closure until now. Slightly different

dose regimens, varying between 0.1 and 0.2 mg/kg every 24-hours and usually for

three doses have been used to treat PDA (Mezu-Ndubuisi et al., 2012). A

prolonged course (> 4 doses) has also been used, but a large meta-analysis of 431

infants noticed no difference in PDA closure, re-treatment or ligation rates when

compared to a short course. Furthermore, there was an increased risk for NEC,

RR 1.87 (95% CI 1.1–3.27), in groups in which prolonged indomethacin courses

were administrated (Herrera, Holberton, & Davis, 2007).

Early prophylactic indomethacin has been shown to reduce periventricular or

intraventricular haemorrhage but does not improve later neurodevelopmental

outcome. In a multicentre double-blinded RCT trial, infants born before 29 weeks

of gestation with large PDA (“high-risk group”) were randomised to receive

either indomethacin (n=44) or a placebo (n=48). Infants were screened by

ultrasound and the study drug was started by 12 hours of age (prophylactic

treatment). Infants in the indomethacin group had significantly less pulmonary

haemorrhage (2% vs 21%) and a trend of less IVH (4.5% vs 12.5%), but the latter

was statistically not significant. The infants with small PDA (not randomised

infants) had a lower risk of pulmonary haemorrhage, and 80% of those had

spontaneous PDA closure, indicating that an early and precise diagnosis of ductal

shunt volume is important when making the decision about therapy (Kluckow,

Jeffery, Gill, & Evans, 2014). Indomethacin treatment is associated with renal

impairment (Lin et al., 2017) and gastrointestinal complications; the risk being

Page 29: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

27

even stronger if an infant is simultaneously receiving corticosteroids or

nephrotoxic drugs (e.g, aminoglycosides). In a RCT study of extremely low birth

weight infants with significant PDA, the indomethacin group (n=73) received the

dose 0.2+0.1+0.1 mg/kg every 24 hours as compared with the ibuprofen group

(n=71) which received the dose 10+5+5 mg/kg every 24 hours. Indomethacin was

more effective than ibuprofen at closing PDA (66% vs 49%, p= 0.046). However,

renal side effects were more common in the indomethacin group (e.g, higher

creatinine, lower glomerular filtration, lower urinary output) than in the ibuprofen

group. There was no difference in other neonatal morbidities (BPD, IVH, NEC,

and ROP) or mortality between the groups (Lin et al., 2017).

Ibuprofen

Ibuprofen is a nonselective COX-inhibitor that reduces PG-mediated

vasodilatation (Figure 1). It is administrated intravenously or orally, and various

dosing regimens have been proposed. The standard dose of ibuprofen is given

over three days (three doses): 10 mg/kg/day followed by 5 mg/kg/day at 24-hour

intervals for two days (IV and oral administrations) (Van Overmeire, Touw,

Schepens, Kearns, & van den Anker, 2001). Higher doses comprise 15–20 mg/kg

followed by 7.5–10 mg/kg administered every 12 to 24 hours for a total of three

doses (IV and oral); repeated courses have also been used (Dani et al., 2012;

Mitra et al., 2018).

There is some evidence that orally administered ibuprofen is as effective as

IV ibuprofen for PDA closure (Ohlsson et al., 2015; Oncel & Erdeve, 2016).

When compared, oral ibuprofen was as efficacious as IV indomethacin in closing

PDA, and easier to administrate (Aly et al., 2007). In a meta-analysis (68 RCT

studies with 4,802 infants), the effects of indomethacin, ibuprofen and

paracetamol to close hsPDA were studied. A high dose of oral ibuprofen was

associated with the highest likelihood of hsPDA closure, versus standard doses of

IV ibuprofen or IV indomethacin (Mitra et al., 2018). However, high-dose

ibuprofen presents a higher risk of potential side effects, especially in the most

immature infants. Because of the nonselective mechanism of COX-inhibitors,

ibuprofen can cause side effects that include gastrointestinal bleeding, NEC and

pulmonary hypertension. Risks for gastrointestinal side effects are greater when

ibuprofen is combined with cortisone treatment (Peltoniemi et al., 2005). The risk

for pulmonary arterial hypertension (PAH) following ibuprofen treatment is

highest in low gestational age infants and when prenatal risk factors such as small

Page 30: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

28

for gestational age (SGA), maternal hypertension of pregnancy or

oligohydramnion are present (Kim et al., 2016). However, ibuprofen generally

has a better safety profile when compared to indomethacin (Ohlsson et al., 2015).

2.4.4 Surgical ligation of PDA

Surgical ligation of PDA is effective in definitively interrupting the ductal shunt.

Most premature infants who require surgical treatment for PDA are the most

immature (less than 28 gestational weeks) and the sickest, typically with

hemodynamic instability and respiratory insufficiency. Most have had medical

treatment (NSAID) prior to ligation if there has been no contraindication for

medical treatment, such as IVH (Weisz & Giesinger, 2018). The past 10 years

have seen a reduction in ligation rates due to a more conservative approach of

treating PDA (Benitz & Committee on Fetus and Newborn, American Academy

of Pediatrics, 2016) and the concern of side effects related to the procedure.

However, treatment options vary globally between centres and countries. In the

United States, for example, direct surgical ligation in ELGA infants has been

more frequent than in the United Kingdom (A. Kulkarni, Richards, & Duffy,

2013). There is also great variability between centres: California hospital ligation

numbers range from 0% to 67% (H. C. Lee, Durand, Danielsen, Duenas, &

Powers, 2015).

While effective, ligation has been associated with both short and long-term

adverse neonatal outcomes, including post-ligation cardiac syndrome (PLCS),

vocal fold paralysis, pneumothorax, chylothorax, and later neurosensory

impairment in early childhood (A. F. El-Khuffash, Jain, Weisz, Mertens, &

McNamara, 2014; Gould et al., 2003; Ibrahim et al., 2015; Jhaveri et al., 2010;

Ting et al., 2016; Weisz et al., 2014). The independent role of ligation and its

association with later problems are unclear; because ligated infants are usually the

most immature and the sickest, the causality to adverse outcomes is unsure. Most

ligated immature infants have one or several related morbidities simultaneous

with prematurity, such as respiratory distress syndrome (RDS) (with mechanical

ventilation), IVH, BPD, ROP and NEC, and therefore the independent role of

PDA and its treatment in later morbidity and mortality is not clear, and causality

is difficult to show. Thus, a selection bias may exist in studies that compare

ligated and non-ligated infants and their later outcomes. There are also studies

where ligation (either direct or after failed medication therapy) has not been

associated with increased mortality or neurodevelopmental impairment (NDI)

Page 31: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

29

when compared to conservative management alone (Weisz et al., 2018). In some

studies, mortality rates in ligated group have been associated with a lower risk of

in-hospital mortality (Weisz et al., 2018). On the other hand, surgical ligation in

VLGA infants seemed to increase the risk of BPD, ROP laser treatment, longer

hospital stay, and longer mechanical ventilation when compared to un-ligated

group (Youn, Moon, Lee, Lee, & Sung, 2017).

Different results show the heterogeneity of the issue and the difficulty of

demonstrating cause-consequence of the ligation and later outcomes, especially in

retrospective and epidemiological studies. However, PLCS after ligation has been

associated with both a higher risk of mortality and a risk of neurodevelopmental

problems later. PLCS may worsen hemodynamic instability and oxygenation (A.

F. El-Khuffash et al., 2014). The optimal selection of patients and the timing of

ligation are key to minimising the procedure’s side effects. The careful selection

of patients for PDA ligation and systematic triage has been shown to reduce the

number of PDA ligations (24% of referrals for surgery were cancelled) without

impacting short-term neonatal morbidity (Resende et al., 2016).

2.4.5 Other options for the treatment

Percutaneous or catheter-based closure has been used to treat PDA for more than

50 years in term neonates (Portsmann & Wierny, 1978). However, the success and

safety of this treatment in very preterm infants is unclear, and has not become

treatment of choice. The procedure has potential future value for premature

infants because it may have fewer complications than surgical ligation. Risks

defined in catheter-based closure are acute arterial injury and aortic or left

pulmonary artery (LPA) obstruction (Backes et al., 2016; Zahn et al., 2016). In a

retrospective analysis performed over a 10-year period at a single centre (n=52),

PDA closure in very preterm infants weighing less than 4 kg was successful in 88%

of the infants. In extremely premature infants (n=24) the successful rate for

transcatheter closure of PDA was also 88% (3/24 failed) (Zahn et al., 2016). A

less invasive procedure is an attractive alternative over surgical ligation, but more

studies are still needed.

Page 32: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

30

2.5 Postnatal age for PDA treatment

The optimal postnatal age for PDA closure is unclear. The timing of medical

treatment has ranged from prophylactic and early treatment at the very first

postnatal days to later treatment at several weeks or even months of age.

2.5.1 Prophylactic and early treatment

Prophylactic treatment of PDA refers to closure of the ductus before 12− 24 hours

of life and before the onset of symptoms in all at risk preterm infants. Previous

trials of prophylactic indomethacin and ibuprofen seemed to decrease the risk of

PDA but caused significant side effects and did not decrease overall morbidity

and mortality (Gournay et al., 2004; Kluckow et al., 2014; Schmidt et al., 2006).

Although prophylactic indomethacin has been shown to reduce the rate of severe

(grades 3 and 4) IVH and pulmonary haemorrhage, it has not demonstrated a

reduction in later morbidities such as BPD, ROP, and NEC, and no improvement

in survival (Schmidt et al., 2006). Indomethacin treatment of large PDA (> 1.7

mm) before 12 hours of age reduced the incidence of pulmonary haemorrhage (9%

vs 23%) and the need of later treatment of PDA (20% vs 40%) but had no effect

on the primary outcome of death or adverse cranial ultrasound findings (Kluckow

et al., 2014). A meta-analysis of prophylactic IV indomethacin found no

improvement in long-term neurodevelopmental outcome or mortality (Fowlie,

Davis, & McGuire, 2010; Schmidt et al., 2001). Prophylactic ibuprofen treatment

has also been tested in preterm infants. Unfortunately, the significantly increased

risk of pulmonary hypertension was found to associate with this approach.

Prophylactic ibuprofen treatment has since been abandoned (Gournay et al., 2004).

In conclusion, prophylactic medical treatment for PDA is not recommended

because of its side effects and because it poses no benefit for later morbidity and

mortality (Cotts, 2009; Ohlsson & Shah, 2011).

Prophylactic or early ligation is not considered a valid treatment for the same

reason and has been abandoned. If surgical closure is performed early, when a

premature infant’s hemodynamic system is both fragile and labile, the risks for

complications rise both during and after the operation. Surgical closure of PDA

may induce a rapid increase in previously low cerebral perfusion as a result of

shunting blood to pulmonary circulation. This, in combination with the fragile

vasculature of the germinal matrix and the potential for reperfusion injury, is

likely to predispose the infant to IVH (Soleymani, Khoo, Noori, & Seri, 2015).

Page 33: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

31

One strategy to prevent hsPDA is early treatment to close the duct very soon

after its diagnosis. In this approach, drug treatment is started before 7 days of life.

Because there is no evidence of benefits, this approach is not recommended by

most neonatologists. Early ibuprofen treatment (median age of 3 days) in VLGA

infants did not have any benefits over delayed treatment (Sosenko, Fajardo,

Claure, & Bancalari, 2012). A standard dose of IV ibuprofen therapy within 72

hours of birth in ELGA infants was effective to close the duct but resulted in no

benefits in later outcomes (e.g, NEC, IVH, BPD, and ROP) (Aranda et al., 2009).

2.5.2 Late treatment (symptomatic therapy)

The criteria and postnatal age for late treatment vary, but late treatment refers to

treatment after 7 days of life. This approach allows for possible spontaneous

closure of the open ductus. The treatment is usually started after hsPDA

symptoms appear or if there is a criterion of hemodynamic significance in

ultrasound. Conservative treatment sometimes refers to treatment even after

discharge. Studies have shown that surgical ligation in VLGA infants before 14

days of life increases the rates of ROP laser treatment, BPD, longer hospital stay

and longer mechanical ventilation when compared to the non-ligated group

(Gudmundsdottir et al., 2015; Youn et al., 2017).

2.5.3 Optimal postnatal age for treatment

The hsPDA should be closed, but what is the optimal time for closure? Premature

closure should be avoided, as possible side effects from the treatments are usually

greater than the shunt effect. The current evidence does not support prophylactic

or early (presymptomatic) closure. At the earliest, the ductal shunt should be

closed after it has turned completely left-to-right. In the case of pulmonary

hypertension (right-to-left shunt) or a ductal cross shunt (pulmonary and systemic

pressure of about the same magnitude), closing the duct can lead to right

ventricular failure. The optimal postnatal age for active ductal closing after the

shunt flow has turned remains controversial. Different trials are inconclusive

regarding when to treat the open ductus, as closing the duct has not affected the

outcomes of other severe neonatal morbidities (e.g, severe BPD, ROP, NEC, and

IVH). The majority of hsPDA cases appear in ELGA infants, who frequently

develop moderate-to-large PDA. Premature closure should be avoided because of

the risk for complications from the treatments, and too-late closure can increase

Page 34: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

32

the risk for morbidities associated with left-to-right shunt. A trial of early vs late

ibuprofen treatment without hsPDA resulted in otherwise similar outcomes, but

early ibuprofen treatment was associated with worse respiratory outcome

(Bancalari, 2016; Bancalari & Jain, 2017; Sosenko et al., 2012).

The individual approach may be the most valid approach for treating PDA.

Treatment choice and postnatal age at treatment should be based on a

pathophysiology-based approach, where the clinical signs of hsPDA, ECHO signs

of hsPDA, and the possible side effects of treatments are evaluated and weighed.

The main dilemma of optimal closing time is that even if the duct is actively

closed, causality to morbidities associated both with the ductal shunt and the

therapies is unclear. Because of the heterogeneity of studies and protocols, it is

difficult to show the long-term benefits of the single therapies. Therefore, the

precise optimal time for treatment is challenging to determine. If the ductal shunt

is small and without harm to the infant, it favours conservative management, but

if it is large and causes severe symptoms such as ventilator problems, pulmonary

haemorrhage or hemodynamic lability, it should be closed. Premature and routine

treatment should be avoided; instead an individual approach should be employed

to evaluate the individual compensatory mechanisms of hemodynamics.

2.6 Paracetamol (acetaminophen)

2.6.1 Paracetamol for pain in preterm infants

Paracetamol (acetaminophen or N-acetyl-p-aminophenol, APAP) is an antipyretic

NSAID used to treat mild to moderate pain in premature infants. Like COX -

inhibitors, the drug inhibits the conversion of arachidonic acid (AA) to

prostaglandins (PGE2, PGI2). Paracetamol acts by lowering cyclooxygenase

products, but the precise mechanism of its action is unclear (Lucas, Warner,

Vojnovic, & Mitchell, 2005).

Page 35: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

33

Fig. 1. Ibuprofen, indomethacin, and paracetamol in the arachidonic acid pathway. COX- inhibitors ibuprofen and indomethacin inhibit the cyclooxygenase (COX) part of prostaglandin. Paracetamol inhibits the peroxidase moiety of the enzyme, decreasing prostaglandin synthesis. Both sides must be active in the prostaglandin synthase enzyme to have the catalysing action.

Paracetamol may also inhibit the descending serotonergic pathways and inhibit

prostaglandin synthesis in the central nervous system. It also may have an active

metabolite influence on cannabinoid receptors (Pacifici & Allegaert, 2014). A

simplified model of the effect of paracetamols on the AA pathway is shown in

Figure 1. Unlike ibuprofen and indomethacin, paracetamol acts on the peroxidase

region of the prostaglandin synthase enzyme, resulting in a decrease in PG

synthesis (Anderson, 2008; Veyckemans, Anderson, Wolf, & Allegaert, 2014).

Paracetamol is primarily eliminated by hepatic metabolism, and hepatic

maturation is the key in paracetamol pharmacokinetics. Paracetamol undergoes

both sulfation and glucuronidation, and these nontoxic products are excreted

renally. In neonates, the sulfation of paracetamol predominates. Paracetamol also

undergoes oxidation by cytochrome P450 (CYP) enzymes, predominantly

CYP2E1, to form the reactive N-acetyl-p-benzoquinone-imine (NAPQI), which is

detoxified by conjugation with glutathione (Figure 2) (Cook, Roberts et al., 2016;

Cook, Stockmann et al., 2016). Repeated paracetamol administration has no effect

on glutathione plasma levels in the preterm infant, potentially reflecting good

glutathione stores.

Page 36: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

34

Fig. 2. Paracetamol (acetaminophen) metabolism main pathways. In preterm infants, sulfation predominates, while the proportion of glucuronidation increases later in childhood and adults. Paracetamol is metabolised primarily in the liver into nontoxic* and toxic** products. Nontoxic products have renal excretion, while toxic NAPQI binds to liver proteins and amino acids. NAPQI production is primarily metabolised by the CYP2E1 pathway, and liver toxicity depends on NAPQI levels, not paracetamol itself.

In studies of paracetamol pharmacokinetics of premature infants, the dose of

paracetamol depends of the gestational age, but patient size (weight) is the major

covariate of clearance variance in neonates (Allegaert, Palmer, & Anderson, 2011).

Volume distribution in premature infants at 27 gestational weeks is 0.64 L/Kg,

reaching the mature value at about 6 months of age (0.4–0.45 L/kg). The

increased volume of distribution in neonates supports the use of a larger initial

dose (loading dose) of 20 mg/kg intravenously to attain paracetamol threshold

concentration sooner (10−20 mg/L) (Veyckemans et al., 2014; Wang et al., 2013).

After the loading dose, the maintenance dose is given, usually a repeated dose of

7.5−10 mg/kg/q6h. The clearance of paracetamol in neonates is one-third of the

mature value reported in adults. Clearance maturation is slow until 40 weeks of

gestation and matures rapidly to reach 90% of adult capacity in the first year of

life (Allegaert et al., 2011; Allegaert, Naulaers, Vanhaesebrouck, & Anderson,

2013; Pacifici & Allegaert, 2014; Wang et al., 2013). However, precise data

Page 37: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

35

regarding the pharmacodynamic of paracetamol in the most immature infants are

still limited.

2.6.2 Paracetamol for patent ductus arteriosus

The use of paracetamol for closure of PDA in preterm infants was first published

in a small case report in 2011 (Hammerman et al., 2011). In that study, oral

paracetamol was given to five premature infants, after which all ducts closed.

Following this promising study, many observational studies were published,

showing that paracetamol could be an alternative for the closure of PDA in

preterm infants (Oncel et al., 2013; Terrin et al., 2016). Thereafter in prospective

RCT studies, paracetamol has been noted to have a closing effect on the DA in

premature infants (Dang et al., 2013; El-Mashad, El-Mahdy, El Amrousy, &

Elgendy, 2017; Oncel et al., 2014). Unlike indomethacin and ibuprofen,

paracetamol has no peripheral vasoconstrictive effect and has fewer side effects

(e.g, gastrointestinal). It would thus provide a more attractive choice for closure

of PDA than indomethacin and ibuprofen.

Primarily, paracetamol is administered to preterm infants intravenously. The

general dose used to treat PDA (15 mg/kg/6q) is greater than that used for pain

and antipyretics (7.5−10 mg/kg/6q). The efficacy of paracetamol has been

compared to COX-inhibitors, and it has been noted to be comparable with

indomethacin and ibuprofen. Both IV and oral paracetamol have been compared

to placebo and COX-inhibitors (IV indomethacin, IV or oral ibuprofen).

Paracetamol has been shown to be as effective as COX-inhibitors ibuprofen and

indomethacin, but with fewer side effects (El-Mashad et al., 2017; Oncel et al.,

2013; Terrin et al., 2016).

Oral and IV paracetamol have been shown to be equally effective (Dang et al.,

2013; Oncel et al., 2014). Although there are challenges in oral route

administration in very preterm infants during their first postnatal days, some

promising studies have shown that oral administration may be a safe and efficient

treatment for PDA (Dang et al., 2013; Oncel & Erdeve, 2016). RCTs have shown

that oral paracetamol is as effective as oral ibuprofen for the treatment of PDA in

infants born preterm, no significant differences between the secondary outcomes

were found (Al-Lawama, Alammori, Abdelghani, & Badran, 2018; Dang et al.,

2013; Ohlsson & Shah, 2015; Oncel et al., 2014). However, paracetamol

metabolism (e.g., peak concentrations and clearance) has been studied mainly in

Page 38: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

36

IV forms. Because enteral paracetamol metabolism differs from the IV route,

dosage and safety of oral paracetamol for PDA need more prospective studies.

Page 39: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

37

Tabl

e 2.

Ran

dom

ised

, con

trol

led

tria

ls o

f par

acet

amol

for p

aten

t duc

tus

arte

riosu

s

Stu

dy

Trea

tmen

t

grou

ps

Stu

dy

desi

gn

Par

acet

amol

dose

Ibu/

indo

dose

Pat

ient

s

(par

a/ib

u/in

do)

Pos

tnat

al

age

drug

star

ted

GW

Res

ults

(duc

tus

clos

ed a

fter f

irst

cour

se) n

(%)

Mai

n

resu

lt

Sid

e ef

fect

s

Dan

g

2013

Po

para

/

Po

ibu

RC

T,

not

blin

ded

15 m

g/kg

/6h

3 da

ys

Ibu

10+5

+5m

g/kg

for 3

day

s

160

(80/

80)

≤14

d ≤3

4 P

ara/

ibu

65(8

1.2%

)/63(

78.8

%)

p=0.

693,

RR

0.8

3 (0

.60−

1.15

)

Par

a as

effe

ctiv

e

as ib

u

Par

a ha

s le

ss

hype

rbili

rubi

nem

ia

and

ge-b

leed

ing

Onc

el

2014

po p

ara

/

po ib

u

RC

T 15

mg/

kg/6

h

3 da

ys

Ibu

10+5

+5m

g/kg

for 3

day

s

90 (4

0/40

)1 48

–96

h ≤3

0 P

ara/

Ibu

29(7

2.5%

)/31/

(77.

5%)

p=0.

600,

RR

0.8

2 (0

.38−

1.76

)

Par

a as

effe

ctiv

e

as ib

u2

No

diffe

renc

e

El-

Mas

had

2016

IV p

ara/

IV

ibu

/ IV

indo

RC

T 15

mg/

kg/6

h

3 da

ys

Ibu

10+5

+5m

g/kg

3 da

ys

indo

0.2

mg/

kg/1

2 h

for

3 do

ses

100/

100/

100

≤14

d <2

8 P

ara/

ibu/

indo

80/

77/8

1

p=0.

868

Par

a as

effe

ctiv

e

as ib

u

and

indo

Par

a ha

s le

ss s

ide

effe

cts

(ren

al,

plat

elet

cou

nt, g

e

blee

ding

)

Yan

g

2016

po p

ara

/

po ib

u

RC

T 15

mg/

kg/6

h

3 da

ys

Ibu

10+5

+5

mg/

kg fo

r 3

days

87 (4

4/43

) 15

h−1

0 d

<37

Par

a/ib

u

31(7

0.5%

)/33/

(76.

7%)p

=0.5

06

Par

a as

effe

ctiv

e

as ib

u

In p

ara

grou

p le

ss

olig

uria

(2.3

%vs

14.0

%)3 ,

ns

p=0.

108

1 Ten

infa

nts

died

(5/5

per

gro

up) b

efor

e co

mpl

etin

g th

e tre

atm

ent.

2 Reo

peni

ng ra

te w

as h

ighe

r in

the

para

cata

mol

gro

up, b

ut w

as n

ot s

tatis

tical

ly s

igni

fican

t

(24.

1% v

s 16

.1%

) p=0

.43.

3 Pla

sma

and

urin

e P

GE

2 lev

els

wer

e lo

wer

afte

r tre

atm

ent i

n th

e ib

upro

fen

grou

p as

com

pare

d to

the

para

ceta

mol

gro

up.

Page 40: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

38

Serum concentrations of paracetamol should be about 10−20 mg/L to have

antipyretic and analgesic effect (Allegaert et al., 2013; Wang et al., 2013).

Therapeutic paracetamol levels for closure of the ductus are unknown. In one

small (n=10) retrospective pilot study of hsPDA closure with enteral paracetamol

treatment (15 mg/kg 4 times/day for 3 days) in premature infants (less than 34

weeks), a higher paracetamol concentration correlated with short-term ductal

response. If the concentration was below 20 mg/L, there was no response in PDA,

showing that the concentration needed to close PDA might be higher than the

target used for pain (Bin-Nun, Fink, Mimouni, Algur, & Hammerman, 2018).

Further studies are needed to define a precise dose of paracetamol used for

treating PDA.

2.6.3 Side effects of paracetamol

Acute and short-term safety of paracetamol is well documented. The drug is

known to have possible acute side effects on the liver. Especially when

administrated in high doses, elevations of liver enzymes have been seen.

Fortunately, the true hepatotoxicity caused by paracetamol is rare in neonates. It is

generated through the production of N-acetyl-p-benzoquinone-imine (NAPQI) by

the hepatic cytochrome P450 (Allegaert et al., 2011; Palmer et al., 2008).

Paracetamol-induced hepatotoxicity is not associated with exposure to the drug

per se but depends more of the amount of exposure to NAPQI and the amount of

protecting agent glutathione stores. In an extreme case report, a premature infant

born at 25 gestational weeks had accidentally received more than a 30- to 60- fold

overdose of paracetamol (446 mg/kg IV). An antidote (a glutathione precursor, N-

acetylcysteine) was given, and the infant survived without side effects (Porta,

Sanchez, Nicolas, Garcia, & Martinez, 2012). One hypothesis for the lack of

acute paracetamol toxicity in preterm infants is that, due to their immaturity, the

production of toxic metabolites (NAPQI) in the liver is low, while the synthesis of

protecting agents such as glutathione in the liver is mature, and thus formation

and stores are good despite prematurity.

Recent studies suggest a potential association between maternal paracetamol

exposures and possible foetal and, later, postnatal side effects for children. Safety

data concerning the long-term side effects of paracetamol use in the neonatal

period are limited. Epidemiological studies have shown that the maternal use of

analgesics in the second trimester or later could be associated with male

reproductive disorders such as cryptorchidism or hypospadias (Fisher et al., 2016;

Page 41: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

39

Kilcoyne & Mitchell, 2017; Snijder et al., 2012). A possible explanation may be

that this affects steroidogenesis and, consequently, results in reduced androgen

(testosterone) production in the foetal testes. In-utero paracetamol side effects

were stronger if exposure occurred during the second trimester and if the mother

was taking the drug for a long term (> 4 weeks). However, the current evidence

regarding foetal exposure to paracetamol and later male reproductive disorders

cannot be concluded directly. The effect of paracetamol use, both in the neonatal

period and later in childhood, on the male testis is unknown.

There is no solid evidence that paracetamol could close the foetal duct when

administered in high (even toxic) doses (Taney, Anastasio, Paternostro, Berghella,

& Roman, 2017). There is one case report of ductal closure in near-term infants

due to maternal self-medication with nimesulide and paracetamol (Simbi,

Secchieri, Rinaldo, Demi, & Zanardo, 2002). The possible association between

paracetamol use during pregnancy and later adverse neurodevelopmental

outcomes has risen in recent studies (Bauer, Kriebel, Herbert, Bornehag, & Swan,

2018). Previous animal studies have shown that the presence of paracetamol,

during the critical period of brain development in mice induced long-lasting

effects on cognitive function (Philippot, Gordh, Fredriksson, & Viberg, 2017;

Viberg, Eriksson, Gordh, & Fredriksson, 2014). However, ibuprofen exposure

during brain growth spurt did not affect brain development in mice (Philippot,

Nyberg, Gordh, Fredriksson, & Viberg, 2016; Philippot et al., 2017). In humans, a

recent review of prenatal paracetamol exposure and later neurodevelopmental

development showed an increased risk of adverse neurodevelopmental outcome.

The association was shown between foetal paracetamol exposure and increased

risk for later ADHD and hyperactivity. The problems with these studies are the

heterogeneity of the protocols and in their outcomes, and therefore the difficulty

to prove causality exists. Paracetamol, which is considered a safe medication for

pain and fever during pregnancy, is the most commonly used medication in

pregnancy (Werler, Mitchell, Hernandez-Diaz, & Honein, 2005). As it is also

widely used in term and preterm infants (Allegaert & van den Anker, 2017),

further research on its safety is urgently needed.

2.7 Association of the treatments of PDA with neonatal morbidities

It is unclear which is worse: the prolonged left-to-right shunt through PDA that

causes circulatory changes or the side effects of the therapies. The strength of the

side effects of NSAIDs medications must be weighed against very rapid surgical

Page 42: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

40

ductal closure and its consequences on the circulation of very preterm

hemodynamically labile and fragile infants (Abdel-Hady, Nasef, Shabaan, & Nour,

2013; Evans & Iyer, 1994; Evans, 2015; Gudmundsdottir et al., 2015; Noori,

2010). The problems of these cause-consequences associations are even more

challenging in extremely premature infants, as later morbidities may simply be

consequences of prematurity itself (Laughon, Simmons, & Bose, 2004).

2.7.1 Associations with bronchopulmonary dysplasia (BPD)

The pathophysiology of BPD is multifactorial. The independent role of PDA and

the causality of its treatments with the risk of BPD is unclear. Previous studies

have suggested that PDA can contribute to the development of BPD by increasing

the fluid filtration to the lungs, thus causing pulmonary oedema and an increased

need for ventilator support (Noori, 2010). A former epidemiological study of

VLGA infants found a slight increase in the odds of BPD in infants with PDA

(Marshall et al., 1999). However, some studies show an association between

severe BPD and treatments for PDA. EPICE, a large European population-based

study (n=6896), indicated that preterm infants (born under 31 weeks) who had

treatment for PDA were at higher risk of BPD or death in all regions around

Europe, with an adjusted RR 1.33 (95% CI 1.18−1.51), than those who were not

treated. Survival without major neonatal morbidity was not related to PDA

treatment, demonstrating that variations in PDA treatments did not explain the

result. The incidence of BPD in 23− to 26-week gestational age infants was lower

in cases of expectant management for PDA than when PDA was treated with

either indomethacin or ligation (Sung et al., 2016). However, in the Trial of

Indomethacin Prophylaxis in Preterms (TIPP) study, the risk of BPD in infants

treated with prophylactic indomethacin was similar when compared to placebo.

Furthermore, in the TIPP study, the incidence of BPD was higher after

prophylactic indomethacin than after placebo treatment in infants without PDA

(Schmidt et al., 2006). Indomethacin implicates oliguria and fluid retention,

potentially predisposing premature infants to lung oedema. This side effect must

be considered when giving NSAID medications to very preterm infants. It has

been shown that sole fluid restriction has no effect in reducing BPD but does have

an effect in reducing PDA. In a systematic review comparing restricted fluid

intake and liberal fluid intake, less fluid was associated with reduction in PDA,

but there was only a trend towards a lower risk of BPD, RR 0.85 (0.63−1.14) (E.

F. Bell & Acarregui, 2008). Surgical ligation (prophylactic, primary, or secondary)

Page 43: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

41

was also associated with an elevated risk of BPD (R. Clyman, Cassady, Kirklin,

Collins, & Philips, 2009; R. I. Clyman, 2013). In summary, active closure of PDA

has not been shown to reduce the risk of BPD, indicating an unclear association

the shunt of PDA and the risk of BPD.

2.7.2 Associations with other neonatal outcomes and mortality

COX-inhibitors have acute side effects, such as oliguria and risk of

gastrointestinal perforation that are described in greater detail in section 2.4.3.

Associations between the treatment of PDA and later neonatal morbidity have

been described. An association has been suggested between ibuprofen and

indomethacin and neonatal morbidities like ROP, NEC, and BPD (R. I. Clyman &

Chorne, 2007; Irmesi, Marcialis, Anker, & Fanos, 2014). An association between

surgical ligation and later neurodevelopmental impairment has been described

(Ibrahim et al., 2015; Sadeck et al., 2014). Surgical ligation after indomethacin

therapy has also been associated with NEC when compared to a conservative

approach (Jhaveri et al., 2010). However, the potential for ELGA infants to

succumb directly because of PDA or its treatments is rare (Patel et al., 2015).

Page 44: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

42

Page 45: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

43

3 Aims of the research Patent ductus arteriosus in preterm infants is one of the very important problems

in neonatal medicine. There are differences in both the definition and the

treatment strategies of hsPDA. COX-inhibitors and the ligation of PDA are

established therapies. In this thesis, we wanted to determine whether paracetamol

has the potential to become a novel treatment for PDA. After observational

studies on the association between paracetamol treatment and the incidence of

PDA, we attempted to explore whether paracetamol treatment for pain has a

biological effect on PDA. The currently available medical therapies and surgical

ligation have side effects in premature infants. We aimed to consolidate and

extend these observations using a national cohort of very preterm infants.

The specific aims of the research were:

– To study, in a retrospective trial, whether the introduction of paracetamol for

the treatment of pain in VLGA infants with respiratory distress would

influence the incidence of PDA (Study I).

– To study in a prospective randomised trial (RCT), whether paracetamol has a

biological effect on the early constriction of open DA in premature infants

(Study II).

– To study the adverse effects of paracetamol used in the early treatment of

preterm infants (II).

– To study the predisposing factors for PDA in VLGA infants in a national

cohort (Study III).

– To study the adverse effects of the therapies used for PDA closure in a large

epidemiological study during the first hospitalisation (Studies III and IV)

– To study the mortality of ELGA infants during the first hospitalisation period

and its possible associations to PDA (Study IV).

Page 46: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

44

Page 47: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

45

4 Patients and methods

4.1.1 Paracetamol for patent ductus arteriosus (I and II)

Study I

This retrospective study consists of 105 VLGA infants and 96 VLGA control

infants born before 32 gestation weeks admitted to the Oulu University Hospital

neonatal intensive care unit (NICU). The paracetamol group infants were born

between June 2009 and December 2011, and controls were born from January

2008 to May 2009. Infants with lethal malformations, chromosomal defects, or

severe congenital infections were excluded. Paracetamol was introduced

originally for VLGA infants’ mild and moderate pain in June 2009. Intravenous

(IV) paracetamol was given to infants before 72 h of age (Perfalgan® 10 mg/ml

solution for infusion, Bristol-Myers Squibb Finland, Espoo, Finland) The first

loading dose of paracetamol was 20 mg/kg, followed by 7.5 mg/kg maintenance

dose every 6 h. Paracetamol was given if there were clinical signs of pain, which

were systematically screened with the neonatal infant acute pain assessment scale

(NIAPAS). If the patient did not improve after starting paracetamol, IV morphine

was available. Paracetamol was stopped when no signs of pain were observed

during 24 h.

Heart echo was performed on postnatal days two to five. PDA criteria

included a significant left-to-right shunt with signs of hemodynamic distress (left-

atrium-to-aorta ratio >1.30, tachycardia or high pulse pressure) and persistent

respiratory distress. The size of PDA was measured as internal ductal calibre vs

pulmonary artery main branch root calibre. PDA was treated with a general

course of IV ibuprofen (10 mg/kg/day, then twice 5 mg/kg/day) which was

repeated once if necessary. Surgical closure of PDA was performed when

ibuprofen was not effective or was contraindicated. The closure of PDA was

determined by cardiac ultrasound. Moderate to severe BPD was diagnosed at 36

weeks of gestation using the oxygen weaning test, IVH was defined based on the

international schedule of Papile with several ultrasound exams, and NEC grade 2

to 3 was defined as described by Bell (M. J. Bell et al., 1978; Papile, Burstein,

Burstein, & Koffler, 1978; Walsh et al., 2004). Accurately recorded data on

paracetamol dosages, ibuprofen, surgical ligation, and known side effects of

paracetamol were obtained from the NICU patient database, Centricity Critical

Page 48: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

46

Care Clinisoft (GE Healthcare Finland, Helsinki, Finland). These data were

reviewed with Crystal Reports XI software (SAP Finland Oy, Espoo, Finland).

Permission to use the hospital databases was given by the administrative chief of

Oulu University Hospital. Statistical analyses were performed using the PASW

Statistics 18 (SPSS Inc., Chicago, IL, USA). Continuous variables were analysed

using the independent samples t-test and dichotomous variables using the two-

tailed Chi-squared test. A multivariate analysis of factors influencing the risk of

PDA was performed by logistic regression. Significance was set at p<0.05.

Study II

This study was a randomised, double-blinded, phase I−II trial (The Premature

Infants’ Paracetamol Study, PreParaS study) with the aim of establishing a new

paracetamol indication in high-risk preterm infants The study group consisted of

VLGA infants (born < 32 gestational weeks) born in the Oulu University Hospital

NICU between September 18, 2013 and January 2, 2015. Infants requiring

intensive care were randomly assigned to the IV paracetamol or the placebo (0.45%

NaCl) group (Figure 3). The study was performed in accordance with Good

Clinical Practice guidelines. The hospital ethics committee and the Finnish

Medicines Agency (Fimea) approved the protocol. The monitoring officer

oversaw the trial arrangements regularly. This trial was assigned to the European

Clinical Trials Database (EudraCT 2013-008142-33) and the ClinicalTrials.gov

registry (NCT01938261).

Page 49: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

47

Fig. 3. Recruitment flow chart in PreParaS study.

Randomisation and intervention

Infants were randomised to either the IV paracetamol or the placebo group (IV

0.45% NaCl). Computed randomisation was done using a 4-block design. To

decrease the risk of significant heterogeneity between cases and controls,

individual treatment strata were defined by gender and immaturity. All staff

(nurses and doctors treating patients) were blinded to the study medication. The

dose of paracetamol used for pain in neonates, included a first loading dose of 20

mg/kg, followed by 7.5 mg/kg every 6 h for 4 days, given in 15-minute infusions.

Page 50: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

48

The study drug was started before 24 hours of age. No other paracetamol was

given before, during, or 2 days after (washout period) the study protocol.

Cardiac ultrasound (ECHO)

The first cardiac ultrasound was performed after recruitment but before the study

drug was started. After the study drug was administrated, ECHO was performed

daily until one day after the study medication was finished. Thereafter, in infants

with open DA, cardiac ultrasound to measure ductus calibre was performed once

or twice a week. All infants were examined at the time of discharge from NICU.

The ECHO was performed by neonatologists (n=3) who were trained by

paediatric cardiologist to enhance the uniformity of evaluations. Altogether, 330

ultrasound examinations were performed to study the infants. An interrater

reliability analysis for the 3 raters was calculated using the Cronbach’s α statistic

to determine consistency among raters. Patients were examined in the supine

position with slight left shoulder recumbences. The recordings were obtained

using Vivid i® (first eight patients) or Vivid S5® (GE Healthcare, Helsinki,

Finland) ultrasound devices with appropriate transducers (12S or 6S). No sedation

was used during the ECHO examinations. In cases where the infant was

distressed, the neonatal nurse soothed the infant, and oral 20% glucose solution

drops were administered if needed. ECHO parameters defined were ductal

internal calibre (mm) from the narrowest point of the duct from the pulmonary

side of the vessel and the left-atrium-to-aorta ratio (LA/Ao) from M-mode. The

smallest diameter of the ductal pulmonary end was measured from the

parasagittal plane of the high left parasternal window (ductal view). Diagnostic

criteria of hsPDA were internal ductal diameter > 50% wider than the left

pulmonary artery, LA/Ao ratio >1.4, and flow patterns showing a large left-to-

right ductal flow shunt.

Outcomes and statistics

The primary outcome was the decrease and closure of the ductus during the

intervention as a function of postnatal age. The secondary outcomes were: LA/Ao

ratio, the age of permanent closure of the ductus, treatment for ductus, the side

effects of paracetamol, neonatal morbidity, and mortality. Paracetamol

concentrations were analysed from serum samples stored at -70 C using the

Paracetamol Assay Kit. All samples were analysed as duplicates. The inter- and

Page 51: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

49

intra-assay coefficients of variation were 18.1% and 14.1%, respectively.

Paracetamol concentrations were stable after prolonged storage.

Adverse effects such as low blood pressure and need of inotropes and laboratory

values like platelets, serum sodium, and bilirubin were recorded. Renal function

was monitored from urinary output (mL/kg/hour). Clinical symptoms of hsPDA

were murmur, prolonged ventilation, increased pulse pressure, cardiomegaly,

active precordium and bounding pulses. If PDA required closure, the decision was

made by a clinician. Options for treatment were ibuprofen and surgical closure.

Statistical analysis of Study II

Sample size assumed a 40% proportional difference between groups. It was

assumed that the ductus calibre would diminish during the study medication

period in 50% of the paracetamol-treated infants and 10% of the controls. In

power 80%, alpha-error 0.05 sample size was calculated to be 48 patients.

Outcomes were analysed as the intention to treat. The Mann-Whitney U-test or

the samples t-test was used for continuous variables and the Chi-squared test for

categorical values. Ductal closure postnatal ages were estimated using the

Kaplan-Meier analysis and the hazard ratios were calculated using the Cox

regression analysis. RANOVA (repeated measures analysis of variance) was used

to assess the daily variation in ductal fluid balance between the groups. Statistical

analysis was performed using IBM SPSS version 22.0 for Microsoft Windows

(IBM Acguires SPSS Inc., Chicago, IL, USA).

4.1.2 Morbidities and mortality associated with PDA treatments (III)

The study data were based on the Finnish Medical Birth Register of preterm

infants born before 32 gestational weeks (22−31). Data consisted originally of

4,143 VLGA infants born during the years 2005−2013 (Figure 4). Exclusion

criteria were lethal congenital malformation, lethal chromosomal defect, or death

before 7 days of postnatal age.

Page 52: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

50

Fig. 4. Flow chart of population-based data

PDA treatments, morbidities and mortality, and other risk factors were recorded

from birth until discharge, or until the age of 42+0 postconceptional weeks. The

final study population consisted of 3,668 infants (Figure 4). Diagnosis of hsPDA

was based on the same clinical and echocardiographic criteria mentioned in the

methods for Study I and Study II. The definition of small for gestational age

(SGA) was based on reference values for normal growth in premature infants in

the Finnish population (Sankilampi, Hannila, Saari, Gissler, & Dunkel, 2013).

The duration of gestation was evaluated by ultrasound examination at an

estimated 16 weeks of gestation. The diagnosis of ROP was based on

international guidelines (International Committee for the Classification of

Retinopathy of Prematurity, 2005), and NEC was diagnosed on the basis of Bell

criteria (M. J. Bell et al., 1978), stages II and III being recorded. IVH grades were

defined as described by Papile (Papile et al., 1978). Severe IVH was defined as

Page 53: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

51

being stage 3 or 4. Mild BPD (grade 1) was diagnosed based on the need for

supplementary oxygen or assisted ventilation at 28 postnatal days. If the infant

required less than 30% of supplementary oxygen at 36 weeks of gestation (±6

days), the oxygen reduction test was performed to define grade 2 (moderate) BPD

(Walsh et al., 2004). Severe BPD (grade 3) was diagnosed if the infant required at

least 30% oxygen or assisted ventilation to reach a saturation level of 90–96% at

the age of 36 gestational weeks (±6 days). Due to incomplete data for grade 2

BPD, only severe BPD (grade 3) was included in the final analysis (Table 3).

Inotrope use was recorded if IV inotropic agents (dopamine, dobutamine, and/or

noradrenaline) were administered. Respiratory support was defined in case of

either invasive ventilation (conventional or high-frequency oscillatory ventilation

[HFOV]) or non-invasive ventilation, which were recorded. Non-invasive

ventilation was recorded when nasal continuous positive airway pressure or a

high-flow nasal cannula was needed. All mortalities during the first

hospitalization were recorded up to postconceptional age 42+0 weeks.

Statistical analysis of Study III

A logistic regression analysis, with and without adjustment for gestational age at

birth (weeks), intrauterine growth (SGA), delivery hospital, and antenatal steroids,

was performed to evaluate the effect of early neonatal morbidity (RDS, need for

inotropes, surfactant therapy or mechanical ventilation) on the risk of PDA

requiring treatment. Multivariate logistic regression, with a forward stepwise

variable selection procedure was used to identify the most effective set of

predictors of early neonatal morbidity. A prior multivariate analysis of the

multicollinearity of the predictors used in the univariate analysis was performed

using a variation inflation factor (VIF). If the VIF of a given predictor was greater

than 3, that predictor was eliminated from the multivariate analysis. A logistic

regression analysis was also performed to evaluate the associations between the

treatments for the closure of PDA and serious morbidities, including severe BPD

(grade 3), ROP grades 3–5, NEC stages 2–3, IVH grades 3–4 and death. These

morbidities were adjusted with respect to antenatal factors (gestational weeks,

SGA, delivery hospital and antenatal steroids) and postnatal factor RDS. In

addition, a logistic regression analysis was performed to compare the risk of

severe IVH in the case of early (< 7 days) and late ligation. The results of the

logistic regression analyses were expressed as odds ratios (ORs), and their 95%

confidence intervals (95% CIs). The differences in mean birth weight and

Page 54: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

52

gestational age between the treatment groups were tested with the Student’s t-test,

and differences among more than two groups were evaluated using an analysis of

variance (ANOVA), with Tukey’s HSD correction method for post hoc

comparisons. Differences in gender distribution were tested by the Chi-square test.

All the statistical analyses were performed with IBM SPSS Statistics for

Windows, (Version 22.0, IBM Corp, Armonk, NY).

4.1.3 National survival analysis of infants born extremely preterm (IV).

The study data were based on the Finnish Medical Birth Register of preterm

infants born 2005−2013 before 28 gestation weeks (22+5 ─ 27+6) without

congenital anomalies. The primary aim of the study was to define the mortality

risks of the most premature infants during the first hospitalisation period. The

original data consisted of 1,371 ELGA infants (born < 28 gestational weeks). Those

infants with lethal malformations or genetic defects were excluded.

Statistical analysis of Study IV

Differences in gender distribution were tested by the Chi square test. The

differences in mean birth weight and gestational age were tested with the

Student’s t-test. Mann-Whitney U-test was used to compare medians of skewed

distributed continuous variables, like postnatal age of death. Standardized Normal

Deviate (SND) test was used to compare the differences in the proportions of

neonatal morbidities and mortality between SGA and non-SGA infants.

Associations of neonatal morbidities with mortality were evaluated using logistic

regression analysis adjusted with antenatal steroids, gestational age at birth and

delivery hospital. Analyses were performed separately on SGA and non-SGA

infants. The results of the logistic regression analyses were expressed as odds

ratios (ORs) with 95% confidence intervals (95% CIs). Postnatal age of demise

was evaluated by Kaplan-Meier survival analysis and differences between non-

SGA and SGA infants were tested using Log-Rank test. All the statistical analyses

were performed with IBM SPSS Statistics for Windows, (Version 25.0, IBM Corp,

Armonk, NY).

Page 55: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

53

5 Results

5.1 Paracetamol for patent ductus arteriosus

Study I

This retrospective study showed that the annual incidence of PDA decreased from

30.7% to 14.7% (p=0.008) after the introduction of paracetamol originally given

for pain management to VLGA infants in the NICU. After paracetamol

administration, there was less medical intervention for PDA (ibuprofen treatment

to 15 paracetamol group infants and to 26 controls, p=0.013). Three paracetamol-

exposed and 7 control infants required surgery (Table 3). No adverse events were

seen. Paracetamol treatment was associated with a lower relative risk for PDA

(RR 0.48, p=0.008, Chi-square) and a lower risk for ibuprofen therapy (RR 0.50,

p=0.013). Rate of PDA closure by ibuprofen was similar in males and females (81%

vs 73%).

Page 56: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

54

Table 3. Characteristics of the groups

Characteristics Paracetamol group

n=102

Control group

n=88

p-value

Birth weight, g (SD) 1201 (379) 1308 (369) 0.051

Gestation weeks (SD) 28.5 (1.9) 29.0 (2.1) 0.09

Surfactant doses (SD) 1.1 (0.9) 0.9 (0.9) 0.21

Ventilation days (SD) 3.3 (7.1) 3.8 (9.4) 0.68

PDA diagnosis, n (%) 15 (14.7) 27 /30.7) 0.008

Ibuprofen treatment, n(%) 15 (14.7) 26 (29.5) 0.013

Surgical ligation, n (%) 3 (2.9) 7 (8.0) 0.12

Ductal closure (postnatal

day)

11 (20) 15 (41) 0.43

Highest bilirubin, umol/l, (SD) 150 (24) 159 (23) 0.018

NEC, n (%) 0 1 (1.1) 0.28

IVH gr 3-4, n (%) 4 (3.9) 2 (2.3) 0.52

BPD, gr 2-3 n (%) 11 (10.8) 5 (5.7) 0.21

The number of paracetamol doses per patient (mean, SD) was 17 (11.8). The

postnatal age (hours, SD) for the first dose was 13.2 (13.7), and the mean duration

of paracetamol (days, SD) therapy was 4.3 (3.1), showing that paracetamol

therapy was begun as a prophylactic drug very early in postnatal life. Short

duration of pregnancy decreased (OR 0.58, 95%CI 0.47−0.71), p<0.001), and

paracetamol increased the odds (OR 4.2, 95%CI 1.8−9.7, p=0.001) of closure of

PDA.

Study II

This randomised, controlled, and double-blinded study protocol showed for the

first time that paracetamol has a biological effect on PDA in very premature

infants soon after birth. The DA closed earlier in the paracetamol group than in

the control group infants (Figure 5). The protocol of serial cardiac ultrasound

examinations (n=330) was performed to all study infants. Interrater reliability (3

examiners) was alpha 0.97 (95%CI 0.90−0.99). Baseline characteristics were

similar in both groups (Table 4).

Page 57: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

55

Table 4. Baseline characteristics of the PreParaS study

Characteristics Paracetamol group (n=23) Placebo group (n=25)

Gestational weeks, wk (SD) 28.4 (2.4) 28.3 (2.1)

Birth weight, kg, mean (SD) 1.22 (0.4) 1.12 (0.3)

Antenatal steroids, n (%) 20 (87) 25 (100)

Antenatal steroids, doses/mother,

mean (SD)

1.65 (0.9) 1.64 (0.5)

PROM, n (%) 10 (43) 6 (24)

Caesarean section, n (%) 15 (65) 10 (40)

Males (%) 13 (57) 14 (56)

Apgar at 5 min age, median (range) 7 (4-10) 6 (2-9)

Surfactant, n (%) 16 (70) 22 (88)

Surfactant doses per patient, mean (SD) 1.3 (1.3) 1.4 (0.9)

There was no difference in the basic (first measurement) ductal size between

groups. The mean (SD) first calibre of internal ductal diameter was 1.57 (0.7) mm

in the paracetamol group and 1.4 (0.8) mm in the placebo group (P=0.38). In the

paracetamol group, the ductus closed faster at the end of the intervention as

compared to the placebo (0.45% NaCl) group. The number of infants with a

closed DA was higher in the paracetamol group than in the placebo group (HR

0.49, 95% CI 0.25−0.97, P=0.016. The median (IQR) postnatal age (hours) for the

observed ductal closure in the paracetamol group was 41 (33−85), and in the

placebo group 78 (50−375), p=0.045. No difference was seen between the LA/Ao

ratios (p= 0.31). None of the infants had PDA treatments during the study drug

administration. Seven infants (14.6%) had therapy for PDA before their discharge

from the NICU. The mean (SD) postnatal age (hours) for ductal closure in infants

born after 27 weeks of gestation was 80 (151) in the paracetamol group vs 322

(514) in the placebo group (p=0.004). In the extremely preterm infants group

(born < 27 gestational weeks) (n=8), the effect of paracetamol for ductal closure

was not seen (p=0.63). Four (50%) of those infants had therapy for PDA (3 in

paracetamol group and 1 in placebo group).

Page 58: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

56

Fig. 5. Percentage of infants with open ductus in function of postnatal age (hours) in paracetamol and placebo groups.

Paracetamol was well tolerated, and no acute side effects were seen. Secondary

outcomes are listed in Table 5. There was no difference in adverse events nor in

neonatal outcomes between the groups. Renal side effects were also not observed.

The urinary output during the first week of life was similar (p=0.102), and no

difference was seen in oliguria (urine output < 1 ml/kg/h) or polyuria (urine

output > 5 ml/kg/h). There was no difference between the groups regarding

phototherapy periods and highest bilirubin levels. Furthermore, no signs of

paracetamol-induced hypotension were seen as the requirement of inotropes were

similar (p=0.74).

Page 59: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

57

Table 5. Secondary outcomes of the PreParaS study

Characteristics Paracetamol group

N=23

Placebo group

N=25

Treatment for PDA, n (%) 4 (17) 3 (12)

Mechanical ventilation,

d median (range)

1 (0-32) 2 (0-39)

Supplemental oxygen, d mean (SD) 20 (24.5) 22.4 (25.0)

Inotropes, n=0/1/2/31 17/7/0/1 16/5/0/3

Hypernatremia (>150 mmol/L), n (%) 5 (22) 12 (48)

Phototherapy periods, n mean (SD) 3.5 (1.7) 3.9 (2.0)

Highest s-bil, µmol/L, mean (SD) 159 (21) 158 (19)

BPD, gr 2-3 0 1 (4)

IVH gr 1-2, n (%) 5 (22) 9 (36)

Severe IVH, gr 3-4 1 (4) 1 (4)

NEC, grade 3, n (%) 0 1 (4)

ROP treated, n (%) 1 (4) 0

Death, n (%) 0 1 (4) 1=inotropes: 0=none, 1=dopamine/dobutamine, 2=dopamine and dobutamine, 3=dopamine, dobutamine

and norepinephrine

Paracetamol concentrations were analysed in serum samples (n=87). No toxic

level was noticed in paracetamol concentrations (range 0−25.2 mg/L), and no

accumulation of drug level was seen during the drug administration period. Mean

(SD) concentration during day one was 9.8 (5.5) mg/L (n=26), and the same on

day four of the drug administration was 11.8 (6.0) mg/L (n=17, P=0.29). No

gender difference was seen in paracetamol levels (boys’ mean (SD) concentration

was 9.2 (6.2), while girls’ mean was 9.0 (5.6) mg/L) (p=0.91). When the most

immature infants were analysed further, those born < 29 gestational weeks did not

differ in their concentrations from those born > 29 gestational weeks (9.4 (5.5) vs

8.9 (6.2) mg/L, respectively, P=0.71. In conclusion, paracetamol concentrations

did not explain the differences in ductal closures.

5.2 Morbidities and mortality associated with PDA treatments (III)

Original cohort data consisted of 4,143 VLGA infants born in Finland during

2005−2013. Most of the infants (91%) were born in university hospitals, and only

9% were born at local or central hospitals. After exclusion, the final data

consisted of 3,668 VLGA infants whose mean gestational age was 29.1 weeks,

and the mean birth weight was 1243 g (Table 6). PDA treatment had been

Page 60: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

58

administered to 30.9% of the infants, and of those, the majority (90.1%) had

received medical treatment for the closure of PDA. Surgical ligation was

performed in 9.9% (362/3668) of the infants. Surgery was the primary treatment

in 112 cases, and 250 infants had both medical and surgical treatment (Table 6).

In ELGA infants (< 28 weeks), the rate of PDA treatment was three times higher

than among infants born at 28 weeks or later (i.e, 619 of 1060 cases required

therapy for PDA; 58.4% vs 19.7%).

Page 61: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

59

Table 6. Final study population and treatments of PDA.

Characteristics No therapy

n=2536

Medical

N=770

Direct

Ligation

N=112

Both

therapies

N=250

Total

N=3668

Gestational age, week, mean

(SD)

29.7 (1.9) 28.3 (2.1) 26.1 (1.9) 26.1 (1.9) 29.1 (2.2)

Birth weight, kg, mean (SD) 1340 (371) 1115 (336) 834 (297) 846 (278) 1243 (390)

SGA, n (%) 350 (13.8) 111 (14.4) 20 (17.9) 27 (10.8) 508 (13.8)

Antenatal steroids, n (%) 2368 (94.9) 719 (94.5) 100 (90.1) 242 (98) 3429 (94.9)

Apgar at 5 min, md1 (range) 8 (6-9) 7 (6-8) 6 (5-7) 6 (5-7) 7 (6-8)

Intubation, n (%) 917 (37.9) 421 (55.5) 105 (95.5) 189 (75.9) 1632 (46.2)

RDS, n (%) 1394 (55.0) 671 (87.1) 108 (96.4) 239 (95.6) 2412 (65.8)

Surfactant therapy 1232 (48.6) 632 (82.1) 102 (91.1) 233 (93.2) 2199 (60.0)

Conventional ventilation,

n(%)

1359 (53.6) 658 (85.5) 111 (99.1) 246 (98.4) 2374 (64.7)

HFOV, n (%) 215 (8.5) 145 (18.8) 75 (67.0) 116 (46.4) 551 (15.0)

Postnatal steroids, n (%) 186 (7.3) 121 (15.7) 69 (61.6) 104 (41.6) 480 (13.1)

1=median

Postnatal age of the PDA treatments was analysed for different treatment groups.

The mean postnatal age (SD) for the first dose of medical treatment was 4.0 (4.1)

days, and that of primary surgical ligation was 13.8 (11.4). Of those who had

medical treatment followed by surgery, the former was at a mean age of 4.1 (4.7)

days of life, and the latter was at 15.7 (12.0). The infants who responded to

medical treatment were born later in gestation (mean of 28.3; difference of means

of 2.1; 95% CI 1.8–2.4; P<0.001) and had a higher birth weight (mean of 1115 g;

difference of means of 336; 95% CI: 227–311; P<0.001) than the nonresponders.

Those infants who underwent primary surgical ligation had a mean gestational

age of 26.2 weeks (SD 1.9) and a mean birth weight of 834 g (SD 297). However,

the infants who did not have PDA therapy had a longer gestation age (29.7 weeks,

SD 1.9) and a higher birth weight (1340 g, SD 371) than those that required

treatment for PDA (P<0.001 for all three post hoc comparisons). No gender

differences were found with regard to the PDA therapies (P=0.212).

Study III stated that RDS and the need for mechanical ventilation were

independently associated with the risk of PDA requiring therapy. The severity of

lung disease, rather than prematurity per se, was associated with the risk of

hsPDA. After adjustment (for gestational weeks, growth restriction, and hospital

of birth), odds were to RDS (OR 1.9 95%CI 1.0–3.5, p=0.042) and mechanical

ventilation (OR 2.0 95%CI 1.1–3.6, p=0.030). In ventilated infants, however, the

Page 62: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

60

ventilation mode HFOV was independently associated with a decreased risk of

PDA after controlling other variables in the model.

The population-based cohort study of VLGA infants also showed that

medical and surgical treatment of PDA was associated with the risk of severe

BPD (OR: 2.1; 95% CI: 1.4−3.0, P<0.001; and OR: 4.0; 95% CI: 2.0–8.2,

P<0.001, respectively). The association was strongest in ELGA infants who

required both medical treatment and surgical closure of PDA (OR: 8.2; 95% CI:

3.5–19.1; P<0.001). Primary surgical ligation additionally increased the risk of

both severe IVH (OR: 4.3; 95% CI: 2.5–7.3; P<0.001) and grade 2-3 NEC (OR:

2.1; 95% CI: 1.2–3.6); P<0.007). However, medical treatment was associated

with lower mortality. Severe ROP (grades 3−5) was not associated significantly

with any treatment for PDA. In conclusion both medical and surgical therapies for

PDA were associated with severe BPD, and primary surgical ligation was

associated with NEC and severe IVH. Furthermore, ligation during the first week

of life was associated with a remarkably increased risk of severe IVH relative to

later ligation (OR: 4.9; 95% CI: 2.5–9.8; P<0.001). Medical treatment itself and

followed by surgery was associated with lower mortality after adjusting for

gestational age, intrauterine growth restriction (SGA), delivery hospital, and

antenatal steroids.

Table 7 shows the associations of PDA treatments with neonatal morbidity

and mortality during the first hospitalisation period. Different treatment groups

are compared to infants with no treatment for PDA, and associations are shown as

odds ratios (ORs) and their 95% confidence intervals (95% CIs). These results are

adjusted for gestational weeks, SGA, delivery hospital, antenatal steroids, and

RDS.

Page 63: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

61

Table 7. VLGA infants’ neonatal morbidity and mortality ORs in different PDA treatment groups (compared to no PDA treatment).

All <28 gestation weeks

Outcome/ treatment OR 95% CI P-value OR 95% CI P-value

Severe ROP

Medical 1.1 (0.5-2.4) 0.797 0.9 (0.4-2.1) 0.752

Ligation 0.6 (0.1-2.9) 0.532 0.3 (0.04-0.2) 0.231

Both therapies 1.5 (0.6-4.0) 0.399 1.5 (0.6-4.1) 0.388

Severe BPD

Medical 2.1 (1.4-3.0) <0.001 2.3 (1.3-4.1) 0.007

Ligation 4.0 (2.0-8.2) <0.001 4.4 (2.0-9.7) <0.001

Both therapies 5.8 (3.1-11.1) <0.001 8.2 (3.5-9.1) <0.001

NEC gr 2-3

Medical 1.3 (0.9-1.8) 0.151 1.3 (0.8-2.1) 0.245

Ligation 2.1 (1.2-3.6) 0.007 2.2 (1.2-3.9) 0.010

Both therapies 1.4 (0.9-2.3) 0.176 1.4 (0.8-2.3) 0.278

Severe IVH

Medical 1.1 (0.7-1.6) 0.817 0.9 (0.5-1.5) 0.058

Ligation 4.3 (2.5-7.3) <0.001 4.4 (2.4-8.1) <0.001

Both therapies 1.3 (0.8-2.2) 0.311 1.2 (0.7-2.1) 0.601

Mortality

Medical 0.5 (0.3-0.9) 0.011 0.4 (0.2-0.8) 0.008

Ligation 1.0 (0.5-1.8) 0.901 0.9 (0.4-1.7) 0.649

Both therapies 0.3 (0.2-0.7) 0.003 0.3 (0.1-0.5) <0.001

5.3 Mortality of infants born extremely preterm (IV)

The mean gestational weeks (SD) of the data was 25.7 (1.6), and the mean birth

weight (SD) was 800 g (225) Treatment of PDA was administered to 47.6% of the

infants (644/1353). Surgical ligation was performed on 22.2% of the infants

(301/1353), and in those, the direct ligation proportion was 7.0%. Median

postnatal age (days) of death in non-SGA infants was 17.6 (range 307), and that in

SGA infants was 47.8 days (range 208), p=0.053. During the third day of life, the

SGA infants’ proportion for death began to differ from that of normal growth

infants, after that, the survival proportion was poorer in SGA infants through the

first hospitalization period (Figure 6). After adjustment for antenatal steroids,

gestational weeks and delivery hospital growth restriction was a clear independent

risk factor for mortality (OR: 4.6; 95% CI: 2.6–8.1; P<0.001).

Page 64: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

62

Fig. 6. SGA and non-SGA survival curve during the first hospitalisation period

RDS [OR 2.3; 95% CI (1.0−5.2), P=0.040] and NEC were also associated with

higher mortality [OR 4.8; 95% CI (2.9−7.7), P<0.001]. Severe IVH was

associated with mortality in non-SGA infants [OR 3.4; 95% CI (1.9−6.0),

P<0.001], but not in SGA infants [OR 3.3; 95%CI (0.5−23.0), P=0.232]. When

studied further, diagnosis of pre-eclampsia was associated with the lower

incidence of severe IVH. Thereafter, maternal pre-eclampsia was associated with

lower mortality during the first week [OR 0.5; 95%CI (0.4−0.8), P=0.004], but

was not associated after one week of survival [OR 0.6; 95%CI (0.3−1.2),

P=0.178].

None of the treatments for PDA associated with the increased mortality of

ELGA infants after adjustment. However, medication therapy [OR 0.5; 95% CI

(0.3−0.99), P=0.045] and combination therapy [OR 0.3; 95% CI (0.1−0.6),

P=0.001] of PDA associated with lower risk of mortality. This association was not

seen in direct surgical ligation group [OR 1.1; 95% CI (0.6−2.2), P=0.736]. There

was clear difference in causes of death between SGA and non-SGA infants. SGA

Page 65: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

63

infants succumbed to lung diseases (RDS, severe BPD), while NEC and severe

IVH were the major causes of death for normal-growth ELGA infants.

Page 66: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

64

Page 67: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

65

6 Discussion The decision to treat PDA in a very preterm infant should be based on

comprehensive clinical and echocardiographic markers of hsPDA. Adequate

diagnosis should be based on clinical symptoms in combination with ECHO

parameters. ECHO criteria of the diagnosis should be based on both the

magnitude of the left-to-right shunt and its individual impact on the patient’s

hemodynamic changes to define the optimal postnatal time for treatment.

Treatment should not harm the patient more than the disease (the open ductus)

itself. Current evidence does not support prophylactic indomethacin, ibuprofen or

surgical ligation. Premature closure of PDA has not been proven to affect later

outcomes, and the risks of side effects of the early treatments are higher when

compared to later treatment. It is important to avoid overdiagnosis and subsequent

overtreatment. However, the optimal anticipation in the treatments timing is

important, which mean diagnosis at the proper time and treating a

hemodynamically open ductus before hemodynamic collapse. In conservative

treatment, watchful waiting with fluid restriction is a good choice when ductal

flow does not harm the infant but the flow pattern and clinical symptoms require

therapeutic attention. Medical treatment is the primary choice, followed by

surgical ligation according to clinicians’ individual choice.

A retrospective study of paracetamol (I) discovered that the incidence of PDA

was apparently decreased with no adverse effects after introduction of

paracetamol for treatment of pain in VLGA infants. In the present RCT, the

infants were treated with a four-day course of paracetamol or placebo. We found

for the first time that paracetamol has a biological effect on the early constriction

of PDA in very preterm infants. Although the precise mechanism of paracetamol

is unclear, it inhibits the peroxidase moiety of the prostaglandin synthase enzyme,

thus decreasing the main ductal vasodilator PGE2 (with PGI2) levels.

Prostaglandin levels drop, and the DA contracts. According to present RCTs and

additional studies (Dang et al., 2013; El-Mashad et al., 2017; Oncel et al., 2014;

Yang, Gao, Ren, Wang, & Zhang, 2016), there is sufficient evidence to indicate

that paracetamol could be a new medication for closure of PDA in premature

infants. It has been shown to have the same effect as the NSAID drugs ibuprofen

and indomethacin, but with fewer acute side effects on the kidneys and the

gastrointestinal tract. This is because paracetamol does not cause generalised

vasoconstriction as do the COX-inhibitors ibuprofen and indomethacin (Gournay

et al., 2004).

Page 68: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

66

The optimal time for therapeutic closure of PDA in very premature infants is

controversial. Premature closure with either medication or ligation has been

associated with adverse effects in several studies, although in theory it would be

an attractive approach. Study II indicated that very early administration of

paracetamol using the dosage effective for the treatment of pain did not result in

obvious adverse effects. In ELGA infants, however, no effect on the DA was

observed; these infants had neither benefits nor adverse effects. This raises some

questions about whether the dose or regimen should be different in the most

immature infants, those born under 28 weeks. Study II also showed that male

infants responded better to paracetamol than females, but possibly because of the

small sample size, this result was not statistically significant. The sample size of

Study II was able to prove evidence that paracetamol has an acute effect to PDA,

but it was not powered enough to show the effects in the subgroups. However, the

study’s findings on efficacy and safety were promising enough to indicate further

studies, including a large randomised trial.

The dose and course regimen for PDA closure are unclear, especially for the

most immature infants. The effect of paracetamol might be dose dependent, and

serum concentrations required for PDA closure might be higher than required for

the treatment of pain and antipyretics. Paracetamol is a safe drug when used in

regular doses, but there is still no evidence regarding adverse side effects, in the

long term. Therefore, it cannot be recommended as a first line-therapy for PDA.

The role of prolonged left-to-right shunt and its consequences for later

outcomes is controversial. There may be bias in studies comparing “standard”

treatments and conservative treatment because groups may differ in origin or

confounding factors (e.g, hospital differences) may make the cause-consequence

analyses difficult or even impossible. Prolonged open ductus is associated with

BPD, but so are the treatments (R. I. Clyman, 2013; Madan et al., 2009; Schmidt

et al., 2006; Terek, Yalaz, Ulger, Koroglu, & Kultursay, 2014). The possible role

of PDA in BPD aetiology is unclear. The precise time postnatally when the ductal

shunt changes from physiological to pathophysiological, thus causing higher

morbidity and mortality for premature infants, is also controversial. The

epidemiological study III showed that all therapies for PDA were associated with

severe BPD. The strongest association was seen in those infants who had surgical

ligation of PDA after failed medical therapy. Indomethacin has been implicated in

oliguria and fluid retention, potentially predisposing premature infants to lung

oedema (Schmidt et al., 2006). Surgical ligation (prophylactic, primary or

secondary) for PDA has shown to associate with an elevated risk of BPD (R.

Page 69: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

67

Clyman et al., 2009; R. I. Clyman, 2013). There were 112 infants in Study III who

underwent direct ligation for PDA. Unfortunately, the reason for choosing direct

ligation was not identifiable in the data. Most of those infants were ELGA and the

association with severe IVH and ligation must be interpreted with caution, as the

order of these events is unclear. Severe IVH in very preterm infants mostly occur

during the first 3-4 days of life, and the risk is highest in ELGA infants (Szpecht,

Szymankiewicz, Nowak, & Gadzinowski, 2016). In some cases, the diagnosis of

severe IVH may be considered a contraindication to medical therapy of PDA.

However, as the risk of IVH was almost five-fold higher in infants with early

ligation compared to late ligation (> 7 days of life), the potential role of early

ligation in the development of IVH is worth considering. Surgical closure of PDA

may cause a rapid increase in cerebral perfusion. This in combination with the

fragile vasculature of the germinal matrix and potential reperfusion injury is

likely to predispose to bleeding. In the Study III surgical ligation was associated

with risk of NEC. This finding is in accordance with that of a previous study,

which found a significantly lower rate of NEC in ELGA infants treated by a

conservative approach in comparison to infants treated with early ligation

following indomethacin therapy (Jhaveri et al., 2010).

Although epidemiological studies III and IV were comprehensive, they have

several limitations. As with all register-based studies the cause-and-effect

relationships are difficult to verify and should be considered only as associations.

A range of hospital-driven, genetic and acquired factors may potentially influence

to the outcome. Due to the variability in diagnosing hemodynamically significant

PDA the diagnosis depends on both the centre and the individual clinician.

Although the results confirm and extend the associations between PDA and life-

threatening morbidities that have been obtained in other studies of smaller

populations, further prospective follow-up studies are needed.

In conclusion, these results show that ductal treatment should always be

based on a precise and comprehensive clinical (individual) evaluation of both of

the risk of the large ductal shunt and possible adverse consequences of hsPDA

treatments. Paracetamol may prove to be particularly suitable treatment for early

closure of ductus as it additionally appears to be safe. However, more prospective

trials are needed to show the optimal treatment, dosage, time, and associations of

the treatments for later outcomes.

Page 70: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

68

Page 71: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

69

7 Conclusions – The incidence of PDA decreased after the start of paracetamol administration

in the NICU.

– IV paracetamol had a favorable effect on DA contraction.

– No acute adverse effects of paracetamol were seen.

– Paracetamol is a potentially safe and effective drug that induced closure of

the DA in very preterm infants. At present, it cannot be recommended for

first-line therapy in treatment of PDA in premature infants. More prospective

studies are needed to determine before high-quality evidence of the dosage

and the long-term side effects.

– Conservative management (watchful waiting) is a valid therapy for PDA

when the open ductus does not harm the patient’s hemodynamic and organ

perfusion. Because of the possible side effects of routine therapies, premature

closure of the DA with currently available drugs, and in particular, routine

early ligation should be avoided.

– Both medical therapy and surgical ligation associated with later risk for

severe BPD.

– Direct ligation very early (before 7 days of life) is associated with severe IVH

and NEC.

– PDA treatments were not associated with higher mortality in ELGA infants.

Page 72: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

70

Page 73: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

71

References Abdel-Hady, H., Nasef, N., Shabaan, A. E., & Nour, I. (2013). Patent ductus arteriosus in

preterm infants: Do we have the right answers? BioMed Research International, 2013, 676192. doi:10.1155/2013/676192 [doi]

Al-Lawama, M., Alammori, I., Abdelghani, T., & Badran, E. (2018). Oral paracetamol versus oral ibuprofen for treatment of patent ductus arteriosus. The Journal of International Medical Research, 46(2), 811-818. doi:10.1177/0300060517722698 [doi]

Allegaert, K. (2013). Paracetamol to close the patent ductus arteriosus: From serendipity toward evidence based medicine. Journal of Postgraduate Medicine, 59(4), 251-252. doi:10.4103/0022-3859.123141; 10.4103/0022-3859.123141

Allegaert, K., Naulaers, G., Vanhaesebrouck, S., & Anderson, B. J. (2013). The paracetamol concentration-effect relation in neonates. Paediatric Anaesthesia, 23(1), 45-50. doi:10.1111/pan.12076; 10.1111/pan.12076

Allegaert, K., Palmer, G. M., & Anderson, B. J. (2011). The pharmacokinetics of intravenous paracetamol in neonates: Size matters most. Archives of Disease in Childhood, 96(6), 575-580. doi:10.1136/adc.2010.204552 [doi]

Allegaert, K., & van den Anker, J. N. (2017). Perinatal and neonatal use of paracetamol for pain relief. Seminars in Fetal & Neonatal Medicine, 22(5), 308-313. doi:S1744-165X(17)30071-9 [pii]

Aly, H., Lotfy, W., Badrawi, N., Ghawas, M., Abdel-Meguid, I. E., & Hammad, T. A. (2007). Oral ibuprofen and ductus arteriosus in premature infants: A randomized pilot study. American Journal of Perinatology, 24(5), 267-270. doi:10.1055/s-2007-976550 [doi]

Aminoshariae, A., & Khan, A. (2015). Acetaminophen: Old drug, new issues. Journal of Endodontics, 41(5), 588-593. doi:http://dx.doi.org/10.1016/j.joen.2015.01.024

Anderson, B. J. (2008). Paracetamol (acetaminophen): Mechanisms of action. Paediatric Anaesthesia, 18(10), 915-921. doi:10.1111/j.1460-9592.2008.02764.x [doi]

Aranda, J. V., Clyman, R., Cox, B., Van Overmeire, B., Wozniak, P., Sosenko, I., . . . Darko, L. (2009). A randomized, double-blind, placebo- controlled trial on intravenous ibuprofen L-lysine for the early closure of nonsymptomatic patent ductus arteriosus within 72 hours of birth in extremely low-birth-weight infants. American Journal of Perinatology, 26(3), 235-246. doi:10.1055/s-0028-1103515

Backes, C. H., Cheatham, S. L., Deyo, G. M., Leopold, S., Ball, M. K., Smith, C. V., . . . Berman, D. P. (2016). Percutaneous patent ductus arteriosus (PDA) closure in very preterm infants: Feasibility and complications. Journal of the American Heart Association, 5(2), 10.1161/JAHA.115.002923. doi:10.1161/JAHA.115.002923 [doi]

Bancalari, E. (2016). Patent ductus arteriosus and short- and long-term respiratory outcomes. American Journal of Perinatology, 33(11), 1055-1057. doi:10.1055/s-0036-1586112 [doi]

Page 74: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

72

Bancalari, E., & Jain, D. (2017). Management of patent ductus arteriosus: Are we looking at the right outcomes? The Journal of Pediatrics, 182, 10-11. doi:S0022-3476(16)31432-9 [pii]

Bauer, A. Z., Kriebel, D., Herbert, M. R., Bornehag, C. G., & Swan, S. H. (2018). Prenatal paracetamol exposure and child neurodevelopment: A review. Hormones and Behavior, doi:S0018-506X(17)30454-3 [pii]

Bell, E. F., & Acarregui, M. J. (2008). Restricted versus liberal water intake for preventing morbidity and mortality in preterm infants. The Cochrane Database of Systematic Reviews, (1):CD000503. doi(1), CD000503. doi:10.1002/14651858.CD000503.pub2 [doi]

Bell, M. J., Ternberg, J. L., Feigin, R. D., Keating, J. P., Marshall, R., Barton, L., & Brotherton, T. (1978). Neonatal necrotizing enterocolitis. therapeutic decisions based upon clinical staging. Annals of Surgery, 187(1), 1-7.

Benitz, W. E. (2010). Treatment of persistent patent ductus arteriosus in preterm infants: Time to accept the null hypothesis? Journal of Perinatology : Official Journal of the California Perinatal Association, 30(4), 241-252. doi:10.1038/jp.2010.3 [doi]

Benitz, W. E. (2012). Patent ductus arteriosus: To treat or not to treat? Archives of Disease in Childhood.Fetal and Neonatal Edition, 97(2), F80-2. doi:10.1136/archdischild-2011-300381 [doi]

Benitz, W. E., & Committee on Fetus and Newborn, American Academy of Pediatrics. (2016). Patent ductus arteriosus in preterm infants. Pediatrics, 137(1), 10.1542/peds.2015-3730. Epub 2015 Dec 15. doi:10.1542/peds.2015-3730 [doi]

Bin-Nun, A., Fink, D., Mimouni, F. B., Algur, N., & Hammerman, C. (2018). Paracetamol serum concentrations in neonates treated enterally for ductal closure: A pilot study. The Journal of Pediatrics, doi:S0022-3476(18)30048-9 [pii]

Blencowe, H., Cousens, S., Oestergaard, M. Z., Chou, D., Moller, A. B., Narwal, R., . . . Lawn, J. E. (2012). National, regional, and worldwide estimates of preterm birth rates in the year 2010 with time trends since 1990 for selected countries: A systematic analysis and implications. Lancet (London, England), 379(9832), 2162-2172. doi:10.1016/S0140-6736(12)60820-4 [doi]

Boghossian, N. S., Geraci, M., Edwards, E. M., & Horbar, J. D. (2018). Morbidity and mortality in small for gestational age infants at 22 to 29 weeks' gestation. Pediatrics, 141(2), 10.1542/peds.2017-2533. Epub 2018 Jan 18. doi:e20172533 [pii]

Bokenkamp, R., DeRuiter, M. C., van Munsteren, C., & Gittenberger-de Groot, A. C. (2010). Insights into the pathogenesis and genetic background of patency of the ductus arteriosus. Neonatology, 98(1), 6-17. doi:10.1159/000262481; 10.1159/000262481

Bonet, M., Cuttini, M., Piedvache, A., Boyle, E. M., Jarreau, P. H., Kollee, L., . . . MOSAIC and EPICE research groups. (2017). Changes in management policies for extremely preterm births and neonatal outcomes from 2003 to 2012: Two population-based studies in ten european regions. BJOG : An International Journal of Obstetrics and Gynaecology, 124(10), 1595-1604. doi:10.1111/1471-0528.14639 [doi]

Page 75: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

73

Brion, L. P., & Campbell, D. E. (1999). Furosemide in indomethacin-treated infants--systematic review and meta-analysis. Pediatric Nephrology (Berlin, Germany), 13(3), 212-218. doi:10.1007/s004670050595 [doi]

Chu, P. Y., Li, J. S., Kosinski, A. S., Hornik, C. P., & Hill, K. D. (2017). Congenital heart disease in premature infants 25-32 weeks' gestational age. The Journal of Pediatrics, 181, 37-41.e1. doi:S0022-3476(16)31101-5 [pii]

Clyman, R., Cassady, G., Kirklin, J. K., Collins, M., & Philips, J. B.,3rd. (2009). The role of patent ductus arteriosus ligation in bronchopulmonary dysplasia: Reexamining a randomized controlled trial. The Journal of Pediatrics, 154(6), 873-876. doi:10.1016/j.jpeds.2009.01.005 [doi]

Clyman, R. I. (2013). The role of patent ductus arteriosus and its treatments in the development of bronchopulmonary dysplasia. Seminars in Perinatology, 37(2), 102-107. doi:10.1053/j.semperi.2013.01.006 [doi]

Clyman, R. I., & Chorne, N. (2007). Patent ductus arteriosus: Evidence for and against treatment. The Journal of Pediatrics, 150(3), 216-219. doi:S0022-3476(07)00010-8 [pii]

Clyman, R. I., Saha, S., Jobe, A., & Oh, W. (2007). Indomethacin prophylaxis for preterm infants: The impact of 2 multicentered randomized controlled trials on clinical practice. The Journal of Pediatrics, 150(1), 46-50.e2. doi:S0022-3476(06)00842-0 [pii]

Clyman, R. I. (2012). Chapter 54 - patent ductus arteriosus in the preterm infant. In C. A. Gleason, & S. U. Devaskar (Eds.), Avery's diseases of the newborn (ninth edition) (pp. 751-761). Philadelpia: W.B. Saunders. doi:http://dx.doi.org/10.1016/B978-1-4377-0134-0.10054-X

Cook, S. F., Roberts, J. K., Samiee-Zafarghandy, S., Stockmann, C., King, A. D., Deutsch, N., . . . van den Anker, J. N. (2016). Population pharmacokinetics of intravenous paracetamol (acetaminophen) in preterm and term neonates: Model development and external evaluation. Clinical Pharmacokinetics, 55(1), 107-119. doi:10.1007/s40262-015-0301-3 [doi]

Cook, S. F., Stockmann, C., Samiee-Zafarghandy, S., King, A. D., Deutsch, N., Williams, E. F., . . . van den Anker, J. N. (2016). Neonatal maturation of paracetamol (acetaminophen) glucuronidation, sulfation, and oxidation based on a parent-metabolite population pharmacokinetic model. Clinical Pharmacokinetics, 55(11), 1395-1411. doi:10.1007/s40262-016-0408-1 [doi]

Cotts, T. (2009). Escalating dose indomethacin for prophylactic closure of patent ductus arteriosus does not improve closure rates and is associated with increased complications. The Journal of Pediatrics, 154(1), 153. doi:10.1016/j.jpeds.2008.10.028 [doi]

Dang, D., Wang, D., Zhang, C., Zhou, W., Zhou, Q., & Wu, H. (2013). Comparison of oral paracetamol versus ibuprofen in premature infants with patent ductus arteriosus: A randomized controlled trial. PloS One, 8(11), e77888. doi:10.1371/ journal.pone.0077888 [doi]

Page 76: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

74

Dani, C., Vangi, V., Bertini, G., Pratesi, S., Lori, I., Favelli, F., . . . Mosca, F. (2012). High-dose ibuprofen for patent ductus arteriosus in extremely preterm infants: A randomized controlled study. Clinical Pharmacology and Therapeutics, 91(4), 590-596. doi:10.1038/clpt.2011.284 [doi]

De Buyst, J., Rakza, T., Pennaforte, T., Johansson, A. B., & Storme, L. (2012). Hemodynamic effects of fluid restriction in preterm infants with significant patent ductus arteriosus. The Journal of Pediatrics, 161(3), 404-408. doi:10.1016/j.jpeds.2012.03.012 [doi]

Edstedt Bonamy, A. K., Gudmundsdottir, A., Maier, R. F., Toome, L., Zeitlin, J., Bonet, M., . . . collaborators from the EPICE Research Group. (2017). Patent ductus arteriosus treatment in very preterm infants: A european population-based cohort study (EPICE) on variation and outcomes. Neonatology, 111(4), 367-375. doi:10.1159/000454798 [doi]

Edstedt Bonamy, A. K., Zeitlin, J., Piedvache, A., Maier, R. F., van Heijst, A., Varendi, H., . . . Epice Research Group. (2018). Wide variation in severe neonatal morbidity among very preterm infants in european regions. Archives of Disease in Childhood.Fetal and Neonatal Edition, doi:fetalneonatal-2017-313697 [pii]

El-Khuffash, A., James, A. T., Corcoran, J. D., Dicker, P., Franklin, O., Elsayed, Y. N., . . . McNamara, P. J. (2015). A patent ductus arteriosus severity score predicts chronic lung disease or death before discharge. The Journal of Pediatrics, 167(6), 1354-1361.e2. doi:10.1016/j.jpeds.2015.09.028 [doi]

El-Khuffash, A. F., Jain, A., & McNamara, P. J. (2013). Ligation of the patent ductus arteriosus in preterm infants: Understanding the physiology. The Journal of Pediatrics, 162(6), 1100-1106. doi:10.1016/j.jpeds.2012.12.094 [doi]

El-Khuffash, A. F., Jain, A., Weisz, D., Mertens, L., & McNamara, P. J. (2014). Assessment and treatment of post patent ductus arteriosus ligation syndrome. The Journal of Pediatrics, 165(1), 46-52.e1. doi:10.1016/j.jpeds.2014.03.048 [doi]

El-Mashad, A. E., El-Mahdy, H., El Amrousy, D., & Elgendy, M. (2017). Comparative study of the efficacy and safety of paracetamol, ibuprofen, and indomethacin in closure of patent ductus arteriosus in preterm neonates. European Journal of Pediatrics, 176(2), 233-240. doi:10.1007/s00431-016-2830-7 [doi]

Evans, N. (2015). Preterm patent ductus arteriosus: A continuing conundrum for the neonatologist? Seminars in Fetal & Neonatal Medicine, 20(4), 272-277. doi:10.1016/j.siny.2015.03.004 [doi]

Evans, N., & Iyer, P. (1994). Assessment of ductus arteriosus shunt in preterm infants supported by mechanical ventilation: Effect of interatrial shunting. The Journal of Pediatrics, 125(5 Pt 1), 778-785. doi:S0022-3476(94)70078-8 [pii]

Evans, N., & Kluckow, M. (1996). Early ductal shunting and intraventricular haemorrhage in ventilated preterm infants. Archives of Disease in Childhood.Fetal and Neonatal Edition, 75(3), F183-6.

Page 77: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

75

EXPRESS Group, Fellman, V., Hellstrom-Westas, L., Norman, M., Westgren, M., Kallen, K., . . . Wennergren, M. (2009). One-year survival of extremely preterm infants after active perinatal care in sweden. Jama, 301(21), 2225-2233. doi:10.1001/ jama.2009.771 [doi]

Fisher, B. G., Thankamony, A., Hughes, I. A., Ong, K. K., Dunger, D. B., & Acerini, C. L. (2016). Prenatal paracetamol exposure is associated with shorter anogenital distance in male infants. Human Reproduction (Oxford, England), 31(11), 2642-2650. doi:dew196 [pii]

Fowlie, P. W., Davis, P. G., & McGuire, W. (2010). Prophylactic intravenous indomethacin for preventing mortality and morbidity in preterm infants. The Cochrane Database of Systematic Reviews, (7):CD000174. doi(7), CD000174. doi:10.1002/14651858.CD000174.pub2 [doi]

Friedman, W. F., Hirschklau, M. J., Printz, M. P., Pitlick, P. T., & Kirkpatrick, S. E. (1976). Pharmacologic closure of patent ductus arteriosus in the premature infant. The New England Journal of Medicine, 295(10), 526-529. doi:10.1056/ NEJM197609022951003 [doi]

Gentile, R., Stevenson, G., Dooley, T., Franklin, D., Kawabori, I., & Pearlman, A. (1981). Pulsed doppler echocardiographic determination of time of ductal closure in normal newborn infants. The Journal of Pediatrics, 98(3), 443-448.

Gould, D. S., Montenegro, L. M., Gaynor, J. W., Lacy, S. P., Ittenbach, R., Stephens, P., . . . Nicolson, S. C. (2003). A comparison of on-site and off-site patent ductus arteriosus ligation in premature infants. Pediatrics, 112(6 Pt 1), 1298-1301.

Gournay, V., Roze, J. C., Kuster, A., Daoud, P., Cambonie, G., Hascoet, J. M., . . . Thiriez, G. (2004). Prophylactic ibuprofen versus placebo in very premature infants: A randomised, double-blind, placebo-controlled trial. Lancet (London, England), 364(9449), 1939-1944. doi:S014067360417476X [pii]

Gudmundsdottir, A., Johansson, S., Håkansson, S., Norman, M., Källen, K., & Bonamy, A. -. (2015). Timing of pharmacological treatment for patent ductus arteriosus and risk of secondary surgery, death or bronchopulmonary dysplasia: A population-based cohort study of extremely preterm infants. Neonatology, 107(2), 87-92.

Hammerman, C., Bin-Nun, A., Markovitch, E., Schimmel, M. S., Kaplan, M., & Fink, D. (2011). Ductal closure with paracetamol: A surprising new approach to patent ductus arteriosus treatment. Pediatrics, 128(6), e1618-21. doi:10.1542/peds.2011-0359 [doi]

Hamrick, S. E., & Hansmann, G. (2010). Patent ductus arteriosus of the preterm infant. Pediatrics, 125(5), 1020-1030. doi:10.1542/peds.2009-3506 [doi]

Herrera, C., Holberton, J., & Davis, P. (2007). Prolonged versus short course of indomethacin for the treatment of patent ductus arteriosus in preterm infants. The Cochrane Database of Systematic Reviews, (2):CD003480. doi(2), CD003480. doi:10.1002/14651858.CD003480.pub3 [doi]

Heymann, M. A., Rudolph, A. M., & Silverman, N. H. (1976). Closure of the ductus arteriosus in premature infants by inhibition of prostaglandin synthesis. The New England Journal of Medicine, 295(10), 530-533. doi:10.1056/ NEJM197609022951004 [doi]

Page 78: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

76

Ibrahim, M. H., Azab, A., Kamal, N. M., Salama, M. A., Elshorbagy, H. H., Abdallah, E. A., . . . Sherief, L. M. (2015). Outcomes of early ligation of patent ductus arteriosus in preterms, multicenter experience. Medicine, 94(25), e915. doi:10.1097/ MD.0000000000000915 [doi]

International Committee for the Classification of Retinopathy of Prematurity. (2005). The international classification of retinopathy of prematurity revisited. Archives of Ophthalmology (Chicago, Ill.: 1960), 123(7), 991-999. doi:123/7/991 [pii]

Irmesi, R., Marcialis, M. A., Anker, J. V. D., & Fanos, V. (2014). Non-steroidal anti-inflammatory drugs (NSAIDs) in the management of patent ductus arteriosus (PDA) in preterm infants and variations in attitude in clinical practice: A flight around the world. Current Medicinal Chemistry, 21(27), 3132-3152.

Jain, A., & Shah, P. S. (2015). Diagnosis, evaluation, and management of patent ductus arteriosus in preterm neonates. JAMA Pediatrics, 169(9), 863-872. doi:10.1001/ jamapediatrics.2015.0987 [doi]

Janz-Robinson, E. M., Badawi, N., Walker, K., Bajuk, B., Abdel-Latif, M. E., & Neonatal Intensive Care Units Network. (2015). Neurodevelopmental outcomes of premature infants treated for patent ductus arteriosus: A population-based cohort study. The Journal of Pediatrics, 167(5), 1025-32.e3. doi:10.1016/j.jpeds.2015.06.054 [doi]

Jhaveri, N., Moon-Grady, A., & Clyman, R. I. (2010). Early surgical ligation versus a conservative approach for management of patent ductus arteriosus that fails to close after indomethacin treatment. The Journal of Pediatrics, 157(3), 381-7, 387.e1. doi:10.1016/j.jpeds.2010.02.062 [doi]

Ketkeaw, K., Thaithumyanon, P., & Punnahitananda, S. (2004). Nephrocalcinosis in very low birth weight infants: A single center experience. Journal of the Medical Association of Thailand = Chotmaihet Thangphaet, 87 Suppl 2, S72-7.

Kilcoyne, K. R., & Mitchell, R. T. (2017). Assessing the impact of in-utero exposures: Potential effects of paracetamol on male reproductive development. Archives of Disease in Childhood, 102(12), 1169-1175. doi:10.1136/archdischild-2016-311374 [doi]

Kim, S. Y., Shin, S. H., Kim, H. S., Jung, Y. H., Kim, E. K., & Choi, J. H. (2016). Pulmonary arterial hypertension after ibuprofen treatment for patent ductus arteriosus in very low birth weight infants. The Journal of Pediatrics, 179, 49-53.e1. doi:S0022-3476(16)30886-1 [pii]

Kluckow, M., & Evans, N. (2000a). Ductal shunting, high pulmonary blood flow, and pulmonary hemorrhage. The Journal of Pediatrics, 137(1), 68-72. doi:S0022-3476(00)39829-8 [pii]

Kluckow, M., & Evans, N. (2000b). Low superior vena cava flow and intraventricular haemorrhage in preterm infants. Archives of Disease in Childhood.Fetal and Neonatal Edition, 82(3), F188-94.

Kluckow, M., Jeffery, M., Gill, A., & Evans, N. (2014). A randomised placebo-controlled trial of early treatment of the patent ductus arteriosus. Archives of Disease in Childhood.Fetal and Neonatal Edition, 99(2), F99-F104. doi:10.1136/archdischild-2013-304695 [doi]

Page 79: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

77

Koch, J., Hensley, G., Roy, L., Brown, S., Ramaciotti, C., & Rosenfeld, C. R. (2006). Prevalence of spontaneous closure of the ductus arteriosus in neonates at a birth weight of 1000 grams or less. Pediatrics, 117(4), 1113-1121. doi:117/4/1113 [pii]

Kong, X., Xu, F., Wu, R., Wu, H., Ju, R., Zhao, X., . . . Feng, Z. (2016). Neonatal mortality and morbidity among infants between 24 to 31 complete weeks: A multicenter survey in china from 2013 to 2014. BMC Pediatrics, 16(1), 174-016-0716-5. doi:10.1186/s12887-016-0716-5 [doi]

Krichenko, A., Benson, L. N., Burrows, P., Moes, C. A., McLaughlin, P., & Freedom, R. M. (1989). Angiographic classification of the isolated, persistently patent ductus arteriosus and implications for percutaneous catheter occlusion. The American Journal of Cardiology, 63(12), 877-880. doi:0002-9149(89)90064-7 [pii]

Kulkarni, A., Richards, J., & Duffy, D. (2013). Survey of management of patent ductus arteriosus in neonatal units across england. Archives of Disease in Childhood.Fetal and Neonatal Edition, 98(5), F465-6. doi:10.1136/archdischild-2013-304317 [doi]

Kulkarni, M., Gokulakrishnan, G., Price, J., Fernandes, C. J., Leeflang, M., & Pammi, M. (2015). Diagnosing significant PDA using natriuretic peptides in preterm neonates: A systematic review. Pediatrics, 135(2), e510-25. doi:10.1542/peds.2014-1995 [doi]

Laughon, M. M., Simmons, M. A., & Bose, C. L. (2004). Patency of the ductus arteriosus in the premature infant: Is it pathologic? should it be treated? Current Opinion in Pediatrics, 16(2), 146-151. doi:10.1097/00008480-200404000-00005

Lee, B. S., Byun, S. Y., Chung, M. L., Chang, J. Y., Kim, H. Y., Kim, E. A., . . . Pi, S. Y. (2010). Effect of furosemide on ductal closure and renal function in indomethacin-treated preterm infants during the early neonatal period. Neonatology, 98(2), 191-199. doi:10.1159/000289206 [doi]

Lee, H. C., Durand, D. J., Danielsen, B., Duenas, G. V., & Powers, R. J. (2015). Hospital variation in medical and surgical treatment of patent ductus arteriosus. American Journal of Perinatology, 32(4), 379-386. doi:10.1055/s-0034-1387931 [doi]

Lin, Y. J., Chen, C. M., Rehan, V. K., Florens, A., Wu, S. Y., Tsai, M. L., . . . Yeh, T. F. (2017). Randomized trial to compare renal function and ductal response between indomethacin and ibuprofen treatment in extremely low birth weight infants. Neonatology, 111(3), 195-202. doi:10.1159/000450822 [doi]

Liu, L., Oza, S., Hogan, D., Chu, Y., Perin, J., Zhu, J., . . . Black, R. E. (2016). Global, regional, and national causes of under-5 mortality in 2000-15: An updated systematic analysis with implications for the sustainable development goals. Lancet (London, England), 388(10063), 3027-3035. doi:S0140-6736(16)31593-8 [pii]

Lucas, R., Warner, T. D., Vojnovic, I., & Mitchell, J. A. (2005). Cellular mechanisms of acetaminophen: Role of cyclo-oxygenase. FASEB Journal : Official Publication of the Federation of American Societies for Experimental Biology, 19(6), 635-637. doi:04-2437fje [pii]

Madan, J. C., Kendrick, D., Hagadorn, J. I., Frantz, I. D.,3rd, & National Institute of Child Health and Human Development Neonatal Research Network. (2009). Patent ductus arteriosus therapy: Impact on neonatal and 18-month outcome. Pediatrics, 123(2), 674-681. doi:10.1542/peds.2007-2781 [doi]

Page 80: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

78

Marshall, D. D., Kotelchuck, M., Young, T. E., Bose, C. L., Kruyer, L., & O'Shea, T. M. (1999). Risk factors for chronic lung disease in the surfactant era: A north carolina population-based study of very low birth weight infants. north carolina neonatologists association. Pediatrics, 104(6), 1345-1350.

McNamara, P. J., & Sehgal, A. (2007). Towards rational management of the patent ductus arteriosus: The need for disease staging. Archives of Disease in Childhood.Fetal and Neonatal Edition, 92(6), F424-7. doi:92/6/F424 [pii]

Mezu-Ndubuisi, O. J., Agarwal, G., Raghavan, A., Pham, J. T., Ohler, K. H., & Maheshwari, A. (2012). Patent ductus arteriosus in premature neonates. Drugs, 72(7), 907-916. doi:10.2165/11632870-000000000-00000 [doi]

Mitra, S., Florez, I. D., Tamayo, M. E., Mbuagbaw, L., Vanniyasingam, T., Veroniki, A. A., . . . Thabane, L. (2018). Association of placebo, indomethacin, ibuprofen, and acetaminophen with closure of hemodynamically significant patent ductus arteriosus in preterm infants: A systematic review and meta-analysis. Jama, 319(12), 1221-1238. doi:10.1001/jama.2018.1896 [doi]

Noori, S. (2010). Patent ductus arteriosus in the preterm infant: To treat or not to treat? Journal of Perinatology : Official Journal of the California Perinatal Association, 30 Suppl, S31-7. doi:10.1038/jp.2010.97 [doi]

Ohlsson, A., & Shah, P. S. (2015). Paracetamol (acetaminophen) for patent ductus arteriosus in preterm or low-birth-weight infants. The Cochrane Database of Systematic Reviews, (3):CD010061. doi(3), CD010061. doi:10.1002/ 14651858.CD010061.pub2 [doi]

Ohlsson, A., & Shah, S. S. (2011). Ibuprofen for the prevention of patent ductus arteriosus in preterm and/or low birth weight infants. Cochrane Database of Systematic Reviews (Online), (7)

Ohlsson, A., Walia, R., & Shah, S. S. (2015). Ibuprofen for the treatment of patent ductus arteriosus in preterm or low birth weight (or both) infants. The Cochrane Database of Systematic Reviews, (2):CD003481. doi(2), CD003481. doi:10.1002/ 14651858. CD003481.pub6 [doi]

Oncel, M. Y., & Erdeve, O. (2016). Oral medications regarding their safety and efficacy in the management of patent ductus arteriosus. World Journal of Clinical Pediatrics, 5(1), 75-81. doi:10.5409/wjcp.v5.i1.75 [doi]

Oncel, M. Y., Yurttutan, S., Degirmencioglu, H., Uras, N., Altug, N., Erdeve, O., & Dilmen, U. (2013). Intravenous paracetamol treatment in the management of patent ductus arteriosus in extremely low birth weight infants. Neonatology, 103(3), 166-169. doi:10.1159/000345337 [doi]

Oncel, M. Y., Yurttutan, S., Erdeve, O., Uras, N., Altug, N., Oguz, S. S., . . . Dilmen, U. (2014). Oral paracetamol versus oral ibuprofen in the management of patent ductus arteriosus in preterm infants: A randomized controlled trial doi:https://doi.org/ 10.1016/j.jpeds.2013.11.008

Pacifici, G. M., & Allegaert, K. (2014). Clinical pharmacology of paracetamol in neonates: A review. Current Therapeutic Research, Clinical and Experimental, 77, 24-30. doi:10.1016/j.curtheres.2014.12.001 [doi]

Page 81: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

79

Palmer, G. M., Atkins, M., Anderson, B. J., Smith, K. R., Culnane, T. J., McNally, C. M., . . . Hunt, R. W. (2008). I.V. acetaminophen pharmacokinetics in neonates after multiple doses. British Journal of Anaesthesia, 101(4), 523-530. doi:10.1093 /bja/aen208 [doi]

Papile, L. A., Burstein, J., Burstein, R., & Koffler, H. (1978). Incidence and evolution of subependymal and intraventricular hemorrhage: A study of infants with birth weights less than 1,500 gm. The Journal of Pediatrics, 92(4), 529-534.

Patel, R. M., Kandefer, S., Walsh, M. C., Bell, E. F., Carlo, W. A., Laptook, A. R., . . . Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. (2015). Causes and timing of death in extremely premature infants from 2000 through 2011. The New England Journal of Medicine, 372(4), 331-340. doi:10.1056/NEJMoa1403489 [doi]

Peltoniemi, O., Kari, M. A., Heinonen, K., Saarela, T., Nikolajev, K., Andersson, S., . . . Hallman, M. (2005). Pretreatment cortisol values may predict responses to hydrocortisone administration for the prevention of bronchopulmonary dysplasia in high-risk infants. The Journal of Pediatrics, 146(5), 632-637. doi:S0022347604012156 [pii]

Perlman, J. M., Hill, A., & Volpe, J. J. (1981). The effect of patent ductus arteriosus on flow velocity in the anterior cerebral arteries: Ductal steal in the premature newborn infant. The Journal of Pediatrics, 99(5), 767-771.

Philippot, G., Gordh, T., Fredriksson, A., & Viberg, H. (2017). Adult neurobehavioral alterations in male and female mice following developmental exposure to paracetamol (acetaminophen): Characterization of a critical period. Journal of Applied Toxicology : JAT, 37(10), 1174-1181. doi:10.1002/jat.3473 [doi]

Philippot, G., Nyberg, F., Gordh, T., Fredriksson, A., & Viberg, H. (2016). Short-term exposure and long-term consequences of neonatal exposure to delta(9)-tetrahydrocannabinol (THC) and ibuprofen in mice. Behavioural Brain Research, 307, 137-144. doi:10.1016/j.bbr.2016.04.001 [doi]

Pladys, P., Beuchee, A., Wodey, E., Treguier, C., Lassel, L., & Betremieux, P. (2001). Patent ductus arteriosus and cystic periventricular leucomalacia in preterm infants. Acta Paediatrica (Oslo, Norway : 1992), 90(3), 309-315.

Porta, R., Sanchez, L., Nicolas, M., Garcia, C., & Martinez, M. (2012). Lack of toxicity after paracetamol overdose in a extremely preterm neonate. European Journal of Clinical Pharmacology, 68(5), 901-902. doi:10.1007/s00228-011-1165-6 [doi]

Portsmann, W., & Wierny, L. (1978). Nonsurgical closure of PDA (10 years experience) (author's transl). [Nechirurgicky uzaver perzistujici arteriove duceje--10 let zkusenosti] Ceskoslovenska Radiologie, 32(4), 217-224.

Reller, M. D., Rice, M. J., & McDonald, R. W. (1993). Review of studies evaluating ductal patency in the premature infant. The Journal of Pediatrics, 122(6), S59-62.

Resende, M. H., More, K., Nicholls, D., Ting, J., Jain, A., & McNamara, P. J. (2016). The impact of a dedicated patent ductus arteriosus ligation team on neonatal health-care outcomes. Journal of Perinatology : Official Journal of the California Perinatal Association, 36(6), 463-468. doi:10.1038/jp.2015.213 [doi]

Page 82: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

80

Roze, J. C., Cambonie, G., Marchand-Martin, L., Gournay, V., Durrmeyer, X., Durox, M., . . . Hemodynamic EPIPAGE 2 Study Group. (2015). Association between early screening for patent ductus arteriosus and in-hospital mortality among extremely preterm infants. Jama, 313(24), 2441-2448. doi:10.1001/jama.2015.6734 [doi]

Sadeck, L. S., Leone, C. R., Procianoy, R. S., Guinsburg, R., Marba, S. T., Martinez, F. E., . . . Alves Filho, N. (2014). Effects of therapeutic approach on the neonatal evolution of very low birth weight infants with patent ductus arteriosus. Jornal De Pediatria, 90(6), 616-623. doi:10.1016/j.jped.2014.04.010 [doi]

Sankilampi, U., Hannila, M. L., Saari, A., Gissler, M., & Dunkel, L. (2013). New population-based references for birth weight, length, and head circumference in singletons and twins from 23 to 43 gestation weeks. Annals of Medicine, 45(5-6), 446-454. doi:10.3109/07853890.2013.803739 [doi]

Schena, F., Francescato, G., Cappelleri, A., Picciolli, I., Mayer, A., Mosca, F., & Fumagalli, M. (2015). Association between hemodynamically significant patent ductus arteriosus and bronchopulmonary dysplasia. The Journal of Pediatrics, 166(6), 1488-1492. doi:10.1016/j.jpeds.2015.03.012 [doi]

Schmidt, B., Davis, P., Moddemann, D., Ohlsson, A., Roberts, R. S., Saigal, S., . . . Trial of Indomethacin Prophylaxis in Preterms Investigators. (2001). Long-term effects of indomethacin prophylaxis in extremely-low-birth-weight infants. The New England Journal of Medicine, 344(26), 1966-1972. doi:10.1056/NEJM200106283442602 [doi]

Schmidt, B., Roberts, R. S., Fanaroff, A., Davis, P., Kirpalani, H. M., Nwaesei, C., & Vincer, M. (2006). Indomethacin prophylaxis, patent ductus arteriosus, and the risk of bronchopulmonary dysplasia: Further analyses from the trial of indomethacin prophylaxis in preterms (TIPP). Journal of Pediatrics, 148(6), 730-734.e1.

Sehgal, A., Paul, E., & Menahem, S. (2013). Functional echocardiography in staging for ductal disease severity : Role in predicting outcomes. European Journal of Pediatrics, 172(2), 179-184. doi:10.1007/s00431-012-1851-0 [doi]

Serenius, F., Sjors, G., Blennow, M., Fellman, V., Holmstrom, G., Marsal, K., . . . EXPRESS study group. (2014). EXPRESS study shows significant regional differences in 1-year outcome of extremely preterm infants in sweden. Acta Paediatrica (Oslo, Norway : 1992), 103(1), 27-37. doi:10.1111/apa.12421 [doi]

Simbi, K. A., Secchieri, S., Rinaldo, M., Demi, M., & Zanardo, V. (2002). In utero ductal closure following near-term maternal self-medication with nimesulide and acetaminophen. Journal of Obstetrics and Gynaecology : The Journal of the Institute of Obstetrics and Gynaecology, 22(4), 440-441. doi:10.1080/01443610220141489 [doi]

Skelton, R., Evans, N., & Smythe, J. (1994). A blinded comparison of clinical and echocardiographic evaluation of the preterm infant for patent ductus arteriosus. Journal of Paediatrics and Child Health, 30(5), 406-411.

Skinner, J., Skinner, J., Alverson, D., & Hunter, S. (2000). Echocardiography for the neonatologist. London: Churchill Livingstone.

Page 83: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

81

Slaughter, J. L., Reagan, P. B., Bapat, R. V., Newman, T. B., & Klebanoff, M. A. (2016). Nonsteroidal anti-inflammatory administration and patent ductus arteriosus ligation, a survey of practice preferences at US children's hospitals. European Journal of Pediatrics, 175(6), 775-783. doi:10.1007/s00431-016-2705-y [doi]

Snijder, C. A., Kortenkamp, A., Steegers, E. A., Jaddoe, V. W., Hofman, A., Hass, U., & Burdorf, A. (2012). Intrauterine exposure to mild analgesics during pregnancy and the occurrence of cryptorchidism and hypospadia in the offspring: The generation R study. Human Reproduction (Oxford, England), 27(4), 1191-1201. doi:10.1093/ humrep/der474 [doi]

Soleymani, S., Khoo, M. C., Noori, S., & Seri, I. (2015). Modeling of neonatal hemodynamics during PDA closure. Conference Proceedings : ...Annual International Conference of the IEEE Engineering in Medicine and Biology Society.IEEE Engineering in Medicine and Biology Society.Annual Conference, 2015, 1886-1889. doi:10.1109/EMBC.2015.7318750 [doi]

Sosenko, I. R., Fajardo, M. F., Claure, N., & Bancalari, E. (2012). Timing of patent ductus arteriosus treatment and respiratory outcome in premature infants: A double-blind randomized controlled trial. The Journal of Pediatrics, 160(6), 929-35.e1. doi:10.1016/j.jpeds.2011.12.031 [doi]

Stephens, B. E., Gargus, R. A., Walden, R. V., Mance, M., Nye, J., McKinley, L., . . . Vohr, B. R. (2008). Fluid regimens in the first week of life may increase risk of patent ductus arteriosus in extremely low birth weight infants. Journal of Perinatology : Official Journal of the California Perinatal Association, 28(2), 123-128. doi:7211895 [pii]

Su, B. H., Watanabe, T., Shimizu, M., & Yanagisawa, M. (1997). Echocardiographic assessment of patent ductus arteriosus shunt flow pattern in premature infants. Archives of Disease in Childhood.Fetal and Neonatal Edition, 77(1), F36-40.

Sung, S. I., Chang, Y. S., Chun, J. Y., Yoon, S. A., Yoo, H. S., Ahn, S. Y., & Park, W. S. (2016). Mandatory closure versus nonintervention for patent ductus arteriosus in very preterm infants. The Journal of Pediatrics, 177, 66-71.e1. doi:http:// dx.doi.org/10.1016/j.jpeds.2016.06.046

Szpecht, D., Szymankiewicz, M., Nowak, I., & Gadzinowski, J. (2016). Intraventricular hemorrhage in neonates born before 32 weeks of gestation-retrospective analysis of risk factors. Child's Nervous System : ChNS : Official Journal of the International Society for Pediatric Neurosurgery, 32(8), 1399-1404. doi:10.1007/s00381-016-3127-x [doi]

Taney, J., Anastasio, H., Paternostro, A., Berghella, V., & Roman, A. (2017). Placental abruption with delayed fetal compromise in maternal acetaminophen toxicity. Obstetrics and Gynecology, 130(1), 159-162. doi:10.1097/AOG.0000000000002089 [doi]

Page 84: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

82

Terek, D., Yalaz, M., Ulger, Z., Koroglu, O. A., & Kultursay, N. (2014). Medical closure of patent ductus arteriosus does not reduce mortality and development of bronchopulmonary dysplasia in preterm infants. Journal of Research in Medical Sciences : The Official Journal of Isfahan University of Medical Sciences, 19(11), 1074-1079.

Terrin, G., Conte, F., Oncel, M. Y., Scipione, A., McNamara, P. J., Simons, S., . . . De Curtis, M. (2016). Paracetamol for the treatment of patent ductus arteriosus in preterm neonates: A systematic review and meta-analysis. Archives of Disease in Childhood.Fetal and Neonatal Edition, 101(2), F127-36. doi:10.1136/archdischild-2014-307312 [doi]

Ting, J. Y., Resende, M., More, K., Nicholls, D., Weisz, D. E., El-Khuffash, A., . . . McNamara, P. J. (2016). Predictors of respiratory instability in neonates undergoing patient ductus arteriosus ligation after the introduction of targeted milrinone treatment. The Journal of Thoracic and Cardiovascular Surgery, 152(2), 498-504. doi:10.1016/j.jtcvs.2016.03.085 [doi]

Tripathi, A., Black, G. B., Park, Y. M., & Jerrell, J. M. (2013). Prevalence and management of patent ductus arteriosus in a pediatric medicaid cohort. Clinical Cardiology, 36(9), 502-506. doi:10.1002/clc.22150 [doi]

Van Bel, F., Van de Bor, M., Stijnen, T., Baan, J., & Ruys, J. H. (1989). Cerebral blood flow velocity changes in preterm infants after a single dose of indomethacin: Duration of its effect. Pediatrics, 84(5), 802-807.

Van Overmeire, B., Touw, D., Schepens, P. J., Kearns, G. L., & van den Anker, J. N. (2001). Ibuprofen pharmacokinetics in preterm infants with patent ductus arteriosus. Clinical Pharmacology and Therapeutics, 70(4), 336-343. doi:S0009-9236(01)13478-8 [pii]

Van Overmeire, B., Van de Broek, H., Van Laer, P., Weyler, J., & Vanhaesebrouck, P. (2001). Early versus late indomethacin treatment for patent ductus arteriosus in premature infants with respiratory distress syndrome. The Journal of Pediatrics, 138(2), 205-211. doi:S0022-3476(01)96421-2 [pii]

Veyckemans, F., Anderson, B. J., Wolf, A. R., & Allegaert, K. (2014). Intravenous paracetamol dosage in the neonate and small infant. British Journal of Anaesthesia, 112(2), 380-381. doi:10.1093/bja/aet559; 10.1093/bja/aet559

Viberg, H., Eriksson, P., Gordh, T., & Fredriksson, A. (2014). Paracetamol (acetaminophen) administration during neonatal brain development affects cognitive function and alters its analgesic and anxiolytic response in adult male mice. Toxicological Sciences : An Official Journal of the Society of Toxicology, 138(1), 139-147. doi:10.1093/toxsci/kft329 [doi]

Vrancken, S. L., van Heijst, A. F., & de Boode, W. P. (2018). Neonatal hemodynamics: From developmental physiology to comprehensive monitoring. Frontiers in Pediatrics, 6, 87. doi:10.3389/fped.2018.00087 [doi]

Page 85: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

83

Walsh, M. C., Yao, Q., Gettner, P., Hale, E., Collins, M., Hensman, A., . . . National Institute of Child Health and Human Development Neonatal Research Network. (2004). Impact of a physiologic definition on bronchopulmonary dysplasia rates. Pediatrics, 114(5), 1305-1311. doi:114/5/1305 [pii]

Wang, C., Allegaert, K., Tibboel, D., Danhof, M., van der Marel, C. D., Mathot, R. A., & Knibbe, C. A. (2013). Population pharmacokinetics of paracetamol across the human age-range from (pre)term neonates, infants, children to adults. Journal of Clinical Pharmacology, doi:10.1002/jcph.259; 10.1002/jcph.259

Weisz, D. E., & Giesinger, R. E. (2018). Surgical management of a patent ductus arteriosus: Is this still an option? Seminars in Fetal & Neonatal Medicine, doi:S1744-165X(18)30042-8 [pii]

Weisz, D. E., Mirea, L., Resende, M. H. F., Ly, L., Church, P. T., Kelly, E., . . . Shah, P. S. (2018). Outcomes of surgical ligation after unsuccessful pharmacotherapy for patent ductus arteriosus in neonates born extremely preterm. The Journal of Pediatrics, doi:S0022-3476(17)31585-8 [pii]

Weisz, D. E., More, K., McNamara, P. J., & Shah, P. S. (2014). PDA ligation and health outcomes: A meta-analysis. Pediatrics, 133(4), e1024-46. doi:10.1542/peds.2013-3431 [doi]

Werler, M. M., Mitchell, A. A., Hernandez-Diaz, S., & Honein, M. A. (2005). Use of over-the-counter medications during pregnancy. American Journal of Obstetrics and Gynecology, 193(3 Pt 1), 771-777. doi:S0002-9378(05)00346-7 [pii]

Yang, B., Gao, X., Ren, Y., Wang, Y., & Zhang, Q. (2016). Oral paracetamol vs. oral ibuprofen in the treatment of symptomatic patent ductus arteriosus in premature infants: A randomized controlled trial. Experimental and Therapeutic Medicine, 12(4), 2531-2536. doi:10.3892/etm.2016.3676 [doi]

Youn, Y., Moon, C. J., Lee, J. Y., Lee, C., & Sung, I. K. (2017). Timing of surgical ligation and morbidities in very low birth weight infants. Medicine, 96(14), e6557. doi:10.1097/MD.0000000000006557 [doi]

Zahn, E. M., Peck, D., Phillips, A., Nevin, P., Basaker, K., Simmons, C., . . . Garg, R. (2016). Transcatheter closure of patent ductus arteriosus in extremely premature newborns: Early results and midterm follow-up. JACC.Cardiovascular Interventions, 9(23), 2429-2437. doi:S1936-8798(16)31545-X [pii]

Zonnenberg, I., & de Waal, K. (2012). The definition of a haemodynamic significant duct in randomized controlled trials: A systematic literature review. Acta Paediatrica (Oslo, Norway : 1992), 101(3), 247-251. doi:10.1111/j.1651-2227.2011.02468.x [doi]

Page 86: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

84

Page 87: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

85

Original publications I Aikio O., Härkin P., Saarela T., & Hallman M. (2014). Early paracetamol treatment

associated with lowered risk of persistent ductus arteriosus in very preterm infants. J Matern Fetal Neonatal Med, 27(12), 1252-6.

II Härkin P., Härmä A., Aikio O., Valkama M., Leskinen M., Saarela T., & Hallman M. (2016). Paracetamol Accelerates Closure of the Ductus Arteriosus after Premature Birth: A Randomized Trial. J Pediatr. 177, 72-77.e2.

III Härkin P., Marttila R., Pokka T., Saarela T., & Hallman M. (2017) Morbidities associated with patent ductus arteriosus in preterm infants. Nationwide cohort study. J Matern Fetal Neonatal Med. 11, 1-8. doi: 10.1080/14767058.2017.1347921.

IV Härkin P., Marttila R., Pokka T., Saarela T., & Hallman M. (2018). National survival analysis of infants born extremely preterm. Manuscript.

Reprinted with permission from Taylor & Francis (I, III) and Elsevier Inc. (II)

Original publications are not included in the electronic version of the dissertation.

Page 88: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

86

Page 89: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

A C T A U N I V E R S I T A T I S O U L U E N S I S

Book orders:Granum: Virtual book storehttp://granum.uta.fi/granum/

S E R I E S D M E D I C A

1467. Tikanmäki, Marjaana (2018) Preterm birth and parental and pregnancy relatedfactors in association with physical activity and fitness in adolescence and youngadulthood

1468. Juvonen-Posti, Pirjo (2018) Work-related rehabilitation for strengthening workingcareers : a multiperspective and mixed methods study of its mechanisms

1469. Palaniswamy, Saranya (2018) Vitamin D status and its association with leukocytetelomere length, obesity and inflammation in young adults : a Northern FinlandBirth Cohort 1966 study

1470. Bur, Hamid (2018) Biological prognostic and predictive markers in Hodgkinlymphoma

1471. Haapanen, Henri (2018) Preconditioning against ischemic injury of the centralnervous system in aortic surgery : an experimental study in a porcine model withremote ischemic preconditioning and diazoxide

1472. Mahlman, Mari (2018) Genetic background and antenatal risk factors ofbronchopulmonary dysplasia

1473. Lantto, Ulla (2018) Etiology and outcome of PFAPA (periodic fever, aphthousstomatitis, pharyngitis and adenitis) syndrome among patients operated withtonsillectomy in childhood

1474. Hintsala, Heidi (2018) Cardiovascular responses to cold exposure in untreatedhypertension

1475. Alakortes, Jaana (2018) Social-emotional and behavioral development problems in1 to 2-year-old children in Northern Finland : reports of mothers, fathers andhealthcare professionals

1476. Puhto, Teija (2018) The burden of healthcare-associated infections in primary andtertiary healthcare wards and the cost of procedure-related prosthetic jointinfections

1477. Honkila, Minna (2018) Chlamydia trachomatis infections in neonates and infants

1478. Krooks, Laura (2018) Malocclusions in relation to facial soft tissue characteristics,facial aesthetics and temporomandibular disorders in the Northern Finland BirthCohort 1966

1479. Hoque Apu, Ehsanul (2018) Migration and invasion pattern analysis of oral cancercells in vitro

Page 90: OULU 2018 D 1480 UNIVERSITY OF OULU P.O. Box 8000 FI …jultika.oulu.fi/files/isbn9789526220253.pdfEarly paracetamol treatment associated with lowered risk of persistent ductus arteriosus

UNIVERSITY OF OULU P .O. Box 8000 F I -90014 UNIVERSITY OF OULU FINLAND

A C T A U N I V E R S I T A T I S O U L U E N S I S

University Lecturer Tuomo Glumoff

University Lecturer Santeri Palviainen

Postdoctoral research fellow Sanna Taskila

Professor Olli Vuolteenaho

University Lecturer Veli-Matti Ulvinen

Planning Director Pertti Tikkanen

Professor Jari Juga

University Lecturer Anu Soikkeli

Professor Olli Vuolteenaho

Publications Editor Kirsti Nurkkala

ISBN 978-952-62-2024-6 (Paperback)ISBN 978-952-62-2025-3 (PDF)ISSN 0355-3221 (Print)ISSN 1796-2234 (Online)

U N I V E R S I TAT I S O U L U E N S I S

MEDICA

ACTAD

D 1480

AC

TAP

ia Härkin

OULU 2018

D 1480

Pia Härkin

CLOSURE OF PATENT DUCTUS ARTERIOSUS IN VERY PRETERM INFANTSPOTENTIAL ROLE OF PARACETAMOL AND CONSEQUENCES OF CURRENT TREATMENTS

UNIVERSITY OF OULU GRADUATE SCHOOL;UNIVERSITY OF OULU,FACULTY OF MEDICINE:MEDICAL RESEARCH CENTER OULU;OULU UNIVERSITY HOSPITAL