ArticlePDF AvailableLiterature Review

Patent Ductus Arteriosus: A Contemporary Perspective for the Pediatric and Adult Cardiac Care Provider

Authors:

Abstract and Figures

The burden of patent ductus arteriosus (PDA) continues to be significant. In view of marked differences in preterm infants versus more mature, term counterparts (viewed on a continuum with adolescent and adult patients), mechanisms regulating ductal patency, genetic contributions, clinical consequences, and diagnostic and treatment thresholds are discussed separately, when appropriate. Among both preterm infants and older children and adults, a range of hemodynamic profiles highlighting the markedly variable consequences of the PDA are provided. In most contemporary settings, transcatheter closure is preferable over surgical ligation, but data on longer‐term outcomes, particularly among preterm infants, are lacking. The present review provides recommendations to identify gaps in PDA diagnosis, management, and treatment on which subsequent research can be developed. Ultimately, the combination of refined diagnostic thresholds and expanded treatment options provides the best opportunities to address the burden of PDA. Although fundamental gaps remain unanswered, the present review provides pediatric and adult cardiac care providers with a contemporary framework in PDA care to support the practice of evidence‐based medicine.
Content may be subject to copyright.
Journal of the American Heart Association
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 1
CONTEMPORARY REVIEW
Patent Ductus Arteriosus: A Contemporary
Perspective for the Pediatric and Adult
Cardiac Care Provider
Carl H. Backes , MD; Kevin D. Hill , MD, MSCI; Elaine L. Shelton , PhD;
Jonathan L. Slaughter , MD, MPH; Tamorah R. Lewis , MD, PhD; Dany E. Weisz , MD;
May Ling Mah , MD; Shazia Bhombal , MD; Charles V. Smith , PhD; Patrick J. McNamara , MD;
William E. Benitz , MD; Vidu Garg , MD
ABSTRACT: The burden of patent ductus ar teriosus (PDA) continues to be significant. In view of marked differences in preterm
infants versus more mature, term counterparts (viewed on a continuum with adolescent and adult patients), mechanisms reg-
ulating ductal patency, genetic contributions, clinical consequences, and diagnostic and treatment thresholds are discussed
separately, when appropriate. Among both preterm infants and older children and adults, a range of hemodynamic profiles
highlighting the markedly variable consequences of the PDA are provided. In most contemporary settings, transcatheter
closure is preferable over surgical ligation, but data on longer- term outcomes, particularly among preterm infants, are lack-
ing. The present review provides recommendations to identify gaps in PDA diagnosis, management, and treatment on which
subsequent research can be developed. Ultimately, the combination of refined diagnostic thresholds and expanded treatment
options provides the best opportunities to address the burden of PDA. Although fundamental gaps remain unanswered, the
present review provides pediatric and adult cardiac care providers with a contemporary framework in PDA care to support the
practice of evidence- based medicine.
Key Words: patent ductus arteriosus pediatric cardiology treatment
The ductus arteriosus (DA) is a vascular structure
that bridges the 2 major arteries leading from the
heart, connecting the proximal descending aorta
to the pulmonary artery near the origin of the left branch
pulmonary artery. The DA is an essential component
of fetal circulation, diverting cardiac output away from
the lungs toward the placenta to support systemic ox-
ygenation. At birth, the placental circulation is clamped
and removed, resistance in the pulmonary vascular
bed decreases, and the lungs become the source of
oxygenation and gas exchange; thus, the DA is no lon-
ger necessary. In normal term infants, the DA closes
in >90% by 48 hours and in 100% by 96 hours of age.1
In preterm infants, structural and physiological imma-
turity of the ductus is associated with later closure of
the DA and increasing the probability that the DA will
remain patent at the equivalent of term gestation.2,3 For
example, among infants born at <26 weeks’ gestation,
the median age at spontaneous DA closure is 66 days,
and at term equivalent age the DA remains patent in
>25%,2 referred to as a patent DA (PDA). In a few indi-
viduals, a PDA will remain open into later childhood or
adult life.
The clinical consequences of a PDA are depen-
dent on its size and the cardiopulmonary status of
the patient. Although fundamental questions on best
treatment practices for the PDA remain unanswered,
the present review is intended to provide pediatric
and adult cardiac care providers with a contemporary
framework to support the practice of evidence- based
Corres ponden ce to: Carl H. Bac kes, MD, Nationwide Children’s Hospital, 700 Children’s Dr, Columbus, OH 43205.
Email: carl.backes@nationwidechildrens.org
This article was sent to Mark W. Russell, MD, Guest Editor, for review by expert referees, editorial decision, and final disposition.
For Sources of Funding and Disclosures, see page 19.
© 2022 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley. This is an open access article under the terms of the Creative
Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use
is non-commercial and no modifications or adaptations are made.
JAHA is available at: www.ahajournals.org/journal/jaha
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 2
Backes et al PDA: A Contemporary Perspective
medicine. In view of marked differences in preterm in-
fants and more mature, term counterparts (viewed on a
continuum with term infants beyond the first months of
life, older children, and adult patients), the mechanisms
regulating ductal patency, genetic contributions, clini-
cal consequences, and diagnostic and treatment ap-
proaches are discussed separately, when appropriate.
NORMAL EMBRYOLOGY, ANATOMY,
AND PHYSIOLOGY
In normal cardiovascular development, the sixth em-
bryonic aortic arches undergo morphological trans-
formations that result in bridges between the main
pulmonary artery and the descending aorta distal to
the left subclavian artery (Figure1).4 The proximal por-
tions of the sixth embryonic arches form the branch
pulmonary arteries, whereas the distal left sixth arch
forms the DA.4 This transformation occurs in the first
8 weeks of fetal development in humans. Although an
essential fetal structure, the DA is abnormal if it remains
patent beyond the neonatal period. The DA may per-
sist in a wide variety of sizes and configurations, with
variable relationships to adjacent structures (Figure2).
These anatomic considerations provide insight into
pathophysiologic consequences (discussed below).
MECHANISMS REGULATING FETAL
DUCTAL PATENCY AND CLOSURE
The fetal DA has intrinsic tone, which requires di-
lating factors to maintain patency in utero (Figure3A).
During early fetal development, endothelially derived
nitric oxide (NO) is the primary relaxing agent and acts
through cyclic guanosine monophosphate (cGMP)/
protein kinase G signaling.5 As the fetus approaches
term gestation, the burden of keeping the DA patent
shifts to prostaglandin E2, originating from the pla-
centa,6 which interacts with the G- protein– coupled
receptor, prostaglandin E2 receptor 4 (EP4), to initiate
the cyclic adenosine monophosphate (cAMP)/protein
kinase A signaling cascade.7 Irrespective of the initiat-
ing signaling molecule, these pathways cooperatively
decrease the concentrations of intracellular calcium,
thereby preventing DA smooth muscle cell contraction
during fetal life.8,9
MECHANISMS REGULATING NORMAL
POSTNATAL DUCTAL CLOSURE
Postnatally, the DA functionally closes and perma-
nently remodels into the fibrous ligamentum arterio-
sum. Several molecular, structural, hemodynamic, and
maternal environmental/infection factors have been
described that contribute to postnatal ductal closure.
Molecular Factors
Postnatal DA closure is facilitated by sharp reductions
in dilating factors and increases in intracellular calcium
levels (Figure3B). The onset of respiration dramatically
Nonstandard Abbreviations and Acronyms
DA ductus arteriosus
HSPDA hemodynamically significant patent
ductus arteriosus
IE infective endocarditis
PAH pulmonary arterial hypertension
RD risk difference
Figure 1. Schematic of embryonic aortic arch system.
The 6 pairs of embryonic aortic arches are shown (left-sided
arches are numbered). Broken-lines represent portions that
involute in normal development. The distal left sixth embryonic
arch normally persists and becomes the PDA, bridging the left
pulmonar y artery to the proximal descending aorta. The right
distal sixth arch normally involutes, as does the eighth segment
of the right dorsal aor ta (*), which results in a leftward aortic
arch. PDA indicates patent ductus arteriosus; LCA, left carotid
arter y; LSCA, left subclavian ar tery; RCA, right carotid ar tery;
and RSCA, right subclavian artery. Reproduced from Schneider
and Moore [4] with permission. Copyright ©2006, American Hear t
Association, Inc.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 3
Backes et al PDA: A Contemporary Perspective
increases the alveolar PaO2, which acts as a principal
regulator of DA closure. Emerging evidence suggests
that immature biochemical oxygen- sensing mecha-
nisms contribute to maintenance of ductal patency in
preterm infants.10
Structural Factors
Compared with term infants, rudimentary or absent
intimal cushions, fewer mature contractile smooth
muscle cells, and lack of vasa vasorum are structural
components that contribute to sustained ductal pa-
tency in preterm infants.
Hemodynamic Factors
Recent evidence suggests PDA tone may also be
regulated by biomechanical factors, particularly
among preterm infants. Although data are mixed,11
some investigators have observed an association
between thrombocytopenia and delayed ductal clo-
sure in very preterm infants.12 Interestingly, among
thrombocytopenic premature infants, platelet transfu-
sions fail to accelerate PDA closure. These observa-
tions have led some health care providers to postulate
that platelet function, not platelet number, may be a
regulator of preterm PDA status.13
Maternal Environmental/Infection Factors
Congenital rubella syndrome may contribute to post-
natal ductal patency.14 Zika virus is emerging as an-
other potential infectious cause of PDA.15 In addition to
infectious origins, prenatal teratogens are associated
with increased incidence of PDA. For example, tetrahy-
drocannabinol concentrations secondary to cannabis
use among pregnant women have been associated
with a greater incidence of PDAs.16,17
GENETIC CONTRIBUTORS
PDA has a complex and multifactorial genetic cause.
Evidence suggests that the PDA is likely 2 overlapping
Figure 2. Variations in patent ductus arteriosus (PDA) configuration.
Illustration of multiple configuration of PDAs: type a (“conical”) ductus, with defined aortic ampulla and constriction near the pulmonary
arter y (PA) end; type b (“window”) ductus, with short length and constriction at the aortic end (wide PA end); type c (“tubular”) ductus,
without constrictions at the aor tic or pulmonary ends; type d (“saccular”) ductus, with constricted aortic and pulmonary ends and a
wide center; type e (“elongated”) ductus, which is narrow with a constricted pulmonary end; and type f (“fetal”) ductus, which is found
largely in premature infants and is long, wide, and tortuous. AO indicates aorta.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 4
Backes et al PDA: A Contemporary Perspective
disorders, wherein preterm PDA arises from structural
and physiological immaturity, and term PDA arises
from genetic alterations.18 PDA is a common finding
in dysmorphic syndromes associated with congenital
heart disease, with an estimated 10% of PDA cases
associated with chromosomal abnormalities (Ta b l e 1).
PDA exists in syndromic and nonsyndromic forms;
syndromic cases of PDA, more common in term in-
fants, are associated with chromosomal aneuploidy
(eg, trisomy 21), chromosomal microdeletion (eg,
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 5
Backes et al PDA: A Contemporary Perspective
22q11.2 deletion), and single- gene defects (eg, Char).
Among term infants, epidemiologic studies report an
increased recurrence risk in siblings of 2% to 4% for
PDA.19 Interestingly, among preterm infants, the inci-
dence of PDA (requiring therapy) in monozygotic twins
is higher than in dizygotic twins.20 However, most cases
of nonsyndromic PDA are attributable to multifactorial
inheritance, wherein underlying genetic predisposi-
tions and environmental triggers at vulnerable times
are likely contributory.21,22 Lack of available human DA
tissues precludes studies on the mechanisms of DA
function, but mouse models have been developed that
provide insight on pathways in DA regulation.23
EPIDEMIOLOGY
PDA represents the most common cardiovascular
condition of preterm infants,24 and the incidence of
PDA is inversely related to gestational age at birth.3
Recent evidence suggests that >50% of infants born
at <26 weeks of gestation have an open ductus be-
yond 2 months postnatal.3 Data among term infants
suggest that PDAs are observed in ≈1 in 2000 births,
accounting for 5% to 10% of all congenital heart dis-
ease.24 Longitudinal cohort studies suggest that the
incidence of “silent” PDA, those cases discovered by
cardiac imaging in the absence of clinical manifesta-
tions, approach 1 in 20 births.25
PATHOPHYSIOLOGY
The hemodynamic consequences of PDA are mark-
edly variable. In infants whose pulmonary vascular re-
sistance decreases at birth and PDA remains patent,
a continuous left- to- right shunt develops. Shunt flow,
according to the Poiseuille Law, is proportional to the
pressure gradient between the aorta and pulmonary
artery and inversely related to the resistance to flow.
The impact of changes in pulmonary and systemic re-
sistances is greater among patients with a larger ductus
and less resistance to flow than in those with a smaller
ductus and greater resistance to flow. Modifiable de-
terminants of pulmonary vascular resistance (eg, PaO2
and pH) also modulate transductal flow.26
A left- to- right ductal shunt results in excessive pul-
monary blood flow. The magnitude of the PDA shunt
and its associated cardiopulmonary interactions dic-
tate the pathophysiologic features of this lesion in
clinical care. Specific features of cardiovascular me-
chanics associated with prematurity predispose
neonates, in particular, to greater PDA- associated
cardiorespiratory compromise. A left- to- right ductal
shunt results in increased pulmonary blood flow and
left heart dilatation, culminating in higher left ventricular
end- diastolic pressures, upstream pulmonary venous
pressure, and pulmonary congestion, a pathophysiol-
ogy exacerbated among preterm neonates because
of greater elastance of immature ventricles.27 Unlike
in adults, in whom pulmonary vascular distention and
capillary bed recruitment assist in managing increased
pulmonary blood flow without associated changes in
capillary hydrostatic pressure, the pulmonary vascular
bed in neonates is already nearly fully recruited and
poorly compliant.28 PDA- associated increases in pul-
monary blood flow therefore result in increased pul-
monary arterial pressure and a shift in the pulmonary
pressure head to downstream capillary filtration sites,
leading to pulmonary interstitial edema, reduced lung
compliance, and impaired oxygenation.29,30 Additional
increases in left atrial and volume and pressure over-
load associated with larger ductal shunts may further
worsen pulmonary venous pressure and culminate in
Figure 3. Molecular pathways involved in int rauterine ductus arteriosus (DA) relaxation (A) and molecular pat hways involved
in postnatal DA constriction (B).
A, Endothelial NO synthase (eNOS)– derived NO, CO, and atrial natriuretic peptide (ANP) initiate the cGMP signaling cascade by
activating membrane - b ound or soluble guanylate cyclase (sGC). cGMP subsequently activates protein kinase G (PKG), which decreases
the intracellular calcium concentration by inhibiting voltage- dependent calcium channels ( VDCCs) and promoting calcium uptake into
the sarcoplasmic reticulum. PKG also activates voltage- gated potassium channel (Kv), ATP- gated potassium channel (KAT P), and large
conductance calcium- activated potassium channel (BKCa), which trigger potassium efflux and membrane hyperpolarization, resulting in
VDCC inhibition. In addition, prostaglandin E2 (PGE2), working through the prostaglandin E2 receptor 4 (EP4), activates adenylyl cyclase.
Adenosine also activates adenylyl cyclase, which, in turn, activates the cAMP/protein kinase A (PKA) signaling cascade. PKA activates
Kv, KATP, and BKCa and inhibits myosin light chain kinase (MLCK), which subsequently reduces phosphor ylation of myosin light chains
(MLCs), thereby preventing vasoconstriction. B, Increased O2 tension increases the concentration of intracellular calcium via several
pathways. First, O2 stimulates mitochondrial production of ATP and H2O2, which inhibit vasodilating KAT P and Kv. H2O2 also activates
the ras homologous protein (Rho)/rho- associated protein kinase (ROCK) cascade, which promotes constriction by inhibiting MLC
phosphatase (MLCP)– mediated MLC dephosphorylation. In addition, O2 upregulates the production of 8- iso- prostaglandin F2α (8- iso-
PGF2α), which signals through thromboxane receptor (TR) to activate Rho/ROCK and inositol trisphosphate (IP3) signaling cascades.
Furthermore, O2 promotes constriction by cytochrome P450 (CYP450)– mediated binding of endothelin- 1 (ET- 1) to endothelin receptor
A (ETA), which subsequently activates the IP3 pathway. Similarly, glutamate activates IP3 signaling via noradrenaline (NA) production.
Once activated, IP3 then binds to IP3 receptor (IP3R) on the sarcoplasmic reticulum, causing movement of calcium into the cytoplasm.
Intracellular calcium concentrations are also regulated by transient receptor potential melastatin- 3 channel (TRPM3), which becomes
activated under hypo- osmotic conditions. Accumulation of intracellular calcium facilitates activation of MLCK, which phosphorylates
MLC, allowing for myosin and actin interaction and subsequent muscle contraction. [Ca2+]i indicates concentration of intracellular
calcium; and [Ca2+]SR, concentration of calcium in the sarcoplasmic reticulum.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 6
Backes et al PDA: A Contemporary Perspective
alveolar edema and further deterioration in respira-
tory mechanics and function. Preterm neonates with
respiratory distress syndrome are more sensitive to
increases in microvascular perfusion pressure and
demonstrate exaggerated increases in interstitial and
alveolar lung fluid accumulation, resulting in second-
ary surfactant dysfunction.31,3 2 Diastolic flow reversal in
the descending aorta attributable to ductal shunting to
the pulmonary artery can occur and is associated with
reduced abdominal organ blood flow.33 Absent or re-
verse end- diastolic flow may occur in the celiac, supe-
rior mesenteric, and middle cerebral arteries, although
only middle cerebral artery flow aberrations have been
associated with neonatal morbidity.34
Table 1. PDA Associated With Genetic Syndromes (Selected)
Syndrome Gene(s) Location Additional cardiac lesions
Tri s om y 2 113 Copies of chromosome 21 Atrioventricular septal defect; atria l
septal de fects; ventr icular septal
defect; tetralogy of Fallot
Tri s om y 1823 Copies of chromosome 18 Atrial and ventr icular s eptal
defects; nonspecific cardiovascular
morphologic abnormalities
Tri s om y 1333 Copies of chromosome 13
22q11.2 deletion4TBX1, CRKL 22q 11. 2 Inter rupted aor tic arch (type
B); tetralogy of Fallot; trun cus
arteriosus; ventricular septal
defects; aortic arch abnormalities
Wolf- Hirschhorn5NSD2, WHSC1, LETM1, CTBP1,
CPL X1, FGFRL1
4p Deletion Atrial and ventricular septal
defects; nonspecific cardiovascular
morphologic abnormalities
Char6TFAP 2B 6 p12. 3 Ventricular septal defect
Cantu7ABCC9, KCNJ8 12p12.1 Bicuspid aortic valve; hypertrophic
cardiomyopathy
Carpenter8RAB23, MEGF8 6p12.1- p11.2, 19q13.2 Nonspecific cardiovascular
morphologic abnormalities
CHARGE9SEMA3E, CHD7 7q 21.11, 8 q1 2. 2 Tetralogy of Fall ot; ventricular
septal defect; atrioventricular septal
defect; aortic arch abnormalities
Holt- Oram10 TBX5 12q 24 .1 Atria l and ventricular septal
defects; hypoplastic left heart
syndrome
Loeys- Dietz (forms 1– 5)11 TGFB R1 (1), TGFBR2 (2), SMAD3
(3), TGFB2 (4), TGFB3 (5)
9q22.33 (1), 3p24.1 (2), 15q22.33 (3),
1q41 (4), 14q24.3 (5)
Aortic aneurysm; aortic dissection;
arterial dissection
Mowa t- Wi lso n12 SMA D1P1, ZEB2 2q22.3 Tetralo gy of Fallot; ve ntricul ar
septal defect
Noonan13 PT P N11 (1), LZ TR1 (2, 10), KRAS
(3), SOS1 (4), RAF1 (5), NRAS
(6), BRAF (7), R IT1 (8 ), SOS2 (9),
MRAS ( 11) , R RAS2 (12 ), M A PK1
(13)
12q24.13 (1), 22q11.21 (2, 10),
12p12.1 (3), 2p22.1 (4), 3p25.2
(5), 1p13.2 (6), 7q34 (7), 1q22 (8),
14q21.3 (9), 3q22.3 (11), 11p15.2
(12), 22q11.22
Dysplastic/stenotic pulmonary
valve; pulm onary arter y stenosis,
atrial septal defect; cardiomyopathy
Periventricular heterotopia (X
linked)14
FLNA Xq28 Bicuspid aortic valve
Rubinste in- Taybi (forms 1 and 2)15 CREBBP (1), EP300 (2) 16p13.3 (1); 22q13.2 (2) Nonspecific cardiovascular
morphologic abnormalities
ABCC9 indicate s ATP- bi nding ca ssette, su bfamily C m ember 9 (ak a, sulfonylurea re ceptor 2) enc oding ge ne; BRAF, B- Raf encoding gene; CH ARGE,
coloboma of the eye, heart defe cts, atres ia of the choanae, retardation of growth and /or developm ent, geni tal hypoplasia, a nd ear abnormalities (eg, de afness);
CHD7, chromodomain– helicase– DNA- binding protein 7 (aka, ATP- dependent helicase CHD7) encoding gene; CPL X1, complex in- 1 enc oding ge ne; CREBBP,
CREB- binding protein encoding gene; CRKL, Crk- like protein enc oding ge ne; CTBP1, C- terminal– binding prote in 1 (aka, CtB P1) encoding ge ne; EP300, histone
acetyltransfe rase p30 0 (aka, p30 0 HAT, adenovirus early regio n 1A– associated p rotein p30 0) encod ing gene; FGFRL1, fibroblast growth factor receptor- like
1 encodi ng gene; FLNA, fi lamin A, α encoding gene; KCNJ8, potassium inwardly rectif ying channel, subfamily J, member 8; K RAS, K- Ras encoding gene;
LETM1, leucine zipper- EF- hand co ntaining transmembrane protein 1 encoding ge ne; LZ TR1, leucine– zipper- like transcriptional regulator 1 encoding gene;
MA PK1, mito gen- activated protein kinase 1 (aka , MAPK1, p42MAPK, ERK2) encoding gene; MEGF8, multiple epidermal growth factor- like domains 8; MRAS,
Ras- re lated protein M- Ra s (aka, mu scle RAS oncoge ne homolog, R- Ras3) encoding gene; NRAS, neuroblastoma RAS viral oncogene homolog encoding gene;
NSD2, nuclear re ceptor binding SE T domain p rotein 2 encoding gene; PDA, patent ductus ar teriosus; PT P N 11, tyrosine- protein phosphatase nonreceptor
type 11 (aka, protein- tyrosine phosphatase 1D, Src homology region 2 domain- containing phosphatase- 2, protein- tyrosine phosphatase 2C) encoding gene;
RAB23, Ras - related protein Rab- 23 encoding ge ne; RA F1, RAF proto- oncogen e serine /threonine- protein kina se (aka, proto- onc ogene c- RAF, c- R af, Raf- 1)
encoding gene; RIT1, GTP- binding prote in Rit1 encoding gene; RRAS2, Ras- related protein R- Ras2 encoding gene; SEMA3E, semaphorin 3E encoding gene;
SMAD, mothers agai nst deca pentap legic ho molog (ak a, SMAD family member) encoding genes; SOS, son of sevenless homolog encoding genes; TBX, T-
box transc ription factor enco ding gen es; TFAP2B, tra nscription factor A P- 2 β (aka, AP2- β) encoding g ene; TGFB, tran sforming growth factor enco ding gen es;
TGFBR, transforming growth factor- β receptor enco ding gen es; WHS C1, probable histone- lysine N- methyltransferase nuclear receptor binding SET domain
protein 2 encoding gene; and ZEB2, zinc finger E- box– binding homeobox 2 encoding gene.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 7
Backes et al PDA: A Contemporary Perspective
DEFINING THE HEMODYNAMICALLY
SIGNIFICANT PDA
Among preterm infants, health care providers are
often unable to clearly discern if the PDA is causal of,
or merely associated with, adverse outcomes. Thus,
contemporary definitions of a hemodynamically signifi-
cant PDA (HSPDA) are not simply echocardiographic-
derived markers of atrial or ventricular chamber
enlargement, but rather various clinical and echocar-
diographic parameters aimed at identifying preterm
infants in whom ductal shunt volumes are estimated
to be primary pathological contributors to physiologic
instability (Ta ble2).35 In contrast, among term infants,
older children, and adults, general consensus is that
echocardiographic evidence of left atrial or left ventric-
ular enlargement attributable to the ductus constitutes
an HSPDA (Tab l e 3).36
CLINICAL CONSEQUENCES IN
PRETERM INFANTS
In the setting of unique developmental vulnerabilities,
PDA among preterm infants is associated with sev-
eral adverse outcomes, including death, that are not
typically observed in older, more mature patients.37- 3 9
Despite decades of investigation, clarity on the relative
contributions to short- and longer- term sequelae of a
persistent ductus, including treatments used to close
the ductus, have not been resolved.40- 43
Respiratory
The potential pathological effects of left- to- right ductal
shunting include an association with increased respiratory
support, mechanical ventilation, and chronic lung disease
(notably, bronchopulmonary dysplasia [BPD]). PDA expo-
sure has been associated with pulmonary hemorrhage,
with estimates suggesting an incidence of 3% to 23%,
depending on the gestational age, ductal size, and as-
sociated PDA treatments.44,45 However, a recent meta-
analysis of early treatment (defined as treatment initiated
by postnatal day 7) compared with expectant manage-
ment for an HSPDA among infants at <37 weeks of ges-
tation observed no differences between the 2 groups in
rates of pulmonary hemorrhage (relative risk [RR], 0.58
[95% CI, 0.30– 1.11]; 5 studies; N=332).46
Emerging data suggest that duration of exposure,
rather than simply the presence or absence of a duc-
tal shunt, may contribute more to observed outcomes.
Among infants born at <28 weeks of gestation, the risks of
the composite outcome of BPD or death were greater after
7 to 13 days of exposure to moderate- to- large PDAs than
in infants who closed their ductus in their first postnatal
week (odds ratio [OR], 2.12 [95% CI, 1.04– 4.32]).47 For the
outcome of increased BPD alone, PDA exposure ≥14 days
was associated with increased BPD (OR, 4.08 [95% CI,
Table 2. Comprehensive Grading Schema for HSPDA Among Preterm Infants
Clinical Echocardiography
Asymptomatic No
PDA
No evidence of ductal flow on 2D or Doppler interrog ation
Mild symptoms
• MAP <8 mmHg (on respiratory support of
NCPAP or mechanical ventilation)
• Feeding intolerance
Small,
non- HSPDA
Transductal d iameter <1.5 mm
Restrictive continuous tra nsduct al flow (DA Vmax >2.0 m/s)
No signs of l eft hea rt volu me loadin g (eg, mitral
regurgi tant jet >2.0 m/s or LA:Ao >1.5:1)
No signs of l eft hea rt pressure loading (eg, E/A ratio
<1.0or IVRT <50)
Moderate symptoms
• MAP 9– 12 mmHg (ventilation requireme nt)
• Evidence of abdom inal distention and/or
persistent emesis
Moderate, HSPDA Transductal d iameter 1.5– 3.0 m m
Unrestr ictive pulsatile transductal flow (DA Vmax <2.0 m/s)
Mild- mod erate lef t hear t volume lo ading (eg, L A:Ao
1. 5 2.1 )
Mild- mod erate lef t hear t pressu re loadin g (eg, E/A ratio
≥1.0 or IVRT 50 60)
Decrea sed or abs ent diastolic flow in the superior
mesenteric, middle cerebral, and/or renal arteries
Severe symptoms
• MAP >12 mmHg (high ventil ation requirements
or HFOV)
• Marked abdominal distention and/or erythema
Large HSPDA Transductal d iameter >3.0 m m
Unrestrictive pulsatile transductal flow
Severe lef t hear t volume loading (eg, LA:Ao >2.1,
mitralregurgitant jet >2.0 m/s)
Severe lef t hear t press ure loadi ng (eg, E/A ratio >1.5 or
IVRT >60)
Reversal of end- diastolic flow in superior mesenteric,
middle cerebral, and/or renal arteries
2D indicate s 2 dimens ional; DA Vmax, transducta l maximal flui d velocit y; E/A (ratio), peak velocit y blood f low from the l eft ventricula r relaxation in ea rly dias tole
(E- wave) to peak veloci ty flow in late diastol e caused by atrial co ntractio n (A- wave); HFOV, high- frequency oscillatory ventilation; HSPDA, hemo dynamically
signif icant PDA; IVRT, isovolumic relaxati on time; LA:Ao, left atrial dia meter to aor tic root diameter rati o; MAP, mean airway pressure; NC PAP, nasal c ontinuous
positive airway pressure; and PDA, patent ductus arteriosus.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 8
Backes et al PDA: A Contemporary Perspective
2.32– 7.22]) compared with infants whose ductus closed
in the first postnatal week.47 However, duration of ductal
patency may be a biomarker of increased pulmonary or
systemic illness, rather than causal, for development or
progression of observed outcomes (BPD or death).48
Neurologic
Independent of the presence of PDA, prematurity is
associated with intraventricular hemorrhage, periven-
tricular leukomalacia, and compromised school- aged
performance.4 9- 51 Most intraventricular hemorrhage oc-
curs in the first postnatal week, coincident with decrease
of pulmonary vascular resistance and emergence of a left-
to- right PDA shunt in some preterm infants. Prophylactic
indomethacin is provided in the first postnatal days and
often before a diagnosis of a PDA. Compared with pla-
cebo, administration of prophylactic indomethacin after
preterm birth reduces the incidence of symptomatic PDA
(risk difference [RD], −0.24 [95% CI, −0.28 to −0.21]; RR,
0.44 [95% CI, 0.38– 0.50]) and surgical PDA ligation (RD,
−0.05 [95% CI, −0.08 to −0.03]; RR, 0.51 [95% CI, 0.37–
0.71] ).52 In addition, prophylactic indomethacin reduces
the incidence of severe periventricular and intraventricu-
lar hemorrhage more effectively than does placebo (RD,
−0.05 [95% CI, −0.07 to −0.02]; RR, 0.66 [95% CI, 0.53–
0.82])52; however, these early neurological benefits may
be unrelated to closure of the ductus.53 However, pro-
phylactic indomethacin is not associated with reduced
mortality or improved neurodevelopment at 18 months of
age (RD, 0.01 [95% CI, −0.04 to 0.06]; RR, 1.02 [95%
CI, 0.90– 1.15]).52,54 In view of these observations, rates of
indomethacin use in the first 24 hours postnatal is ≈7%
across US hospitals.55 In the absence of clear evidence,
controversies persist on the benefits (or lack thereof) and
optimal use of indomethacin prophylaxis for preterm in-
fants.42,56,57,58,59,60 Some investigators have called for the
abandonment of routine prophylactic indomethacin to
all preterm infants,42 whereas others have suggested a
nuanced approach that takes into consideration an in-
dividual center’s baseline risk of intraventricular hemor-
rhage.56 Surgical ductal ligation has been associated with
neurodevelopmental impairment in early childhood,61 but
failure to account for confounding effects because of pa-
tient illness limits data interpretation.62
Intestinal Injury
Diastolic flow reversal in the abdominal aorta and sys-
temic arteries (renal, celiac, and superior mesenteric) is
common among preterm infants with PDA.63 Although
data are mixed, contemporary randomized clinical tri-
als have not observed differences in rates of necrotiz-
ing enterocolitis following ductal closure compared
with nonclosure.64,65 More important, simultaneous
administration of early systemic hydrocortisone and
indomethacin for intraventricular hemorrhage prophy-
laxis increases the risk of spontaneous intestinal per-
foration and is contraindicated.66
CLINICAL CONSEQUENCES AMONG
OLDER PATIENTS (TERM INFANT
THROUGH ADULTHOOD)
Pulmonary Arterial Hypertension and
Eisenmenger Syndrome
Although the precise pathophysiological mechanisms
are not completely understood, long- standing left- to-
right shunting exposes the pulmonary arterial system
Table 3. Comprehensive Grading Schema for HSPDA Among Ol der Children and Adults
PDA size Physiological symptoms
Silent or trivial “Silent” (inaudible) PDAs are asymptomatic*
No hemodynamic or anatomic sequelae
Normal exercise capacity
Normal renal, hepatic, and pulmonary function
Small Small left- to- right shunt, not HSPDA
No restri ctions of exe rcise capacity
Mild/moderate Mild- moderate left- to- right or bidirectional shunt, HSPDA
Mild- moderate hemodynamic or anatomic sequelae (mild/moderate LAE and/or LVE, mild-
moderate left ventricular dysfunction)
Mild or moderate hypoxemia/cyanosis
Mild or moderate PH
Potential for mild rena l, hepatic, a nd pulmonary dysfunction
Large Larg e left- to- right, bidirec tional, or right- to- left shunt
Severe hemodyna mic or anatomic sequ elae (severe LAE and/or LVE, moderate to severe lef t
ventricular dysfunction)
Moderate or severe hypoxemia/cyanosis
Severe PH
Risk of Eisenmeng er syndro me with PH an d right- to- lef t shunting
HSPDA indicates hemodynamically significant PDA, defined as left atrial/ventricular enlargement and/or sustained pulmonary blood flow to systemic blood
flow ratio (Qp/Qs) ≥1.5; LAE, l eft atri al enlargement; LVE, left ventri cular en largem ent; PDA, paten t ductus arterio sus; and PH, pu lmonar y hyper tension.
*Not all asymptomatic PDAs are silent.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 9
Backes et al PDA: A Contemporary Perspective
to higher pressures and greater blood flows. Over time,
this leads to progressive morphological changes in the
pulmonary vasculature, including arteriolar medial hy-
pertrophy, intimal proliferation and fibrosis, and even-
tual obliteration of pulmonary arterioles and capillaries,
characterized by increased pulmonary vascular resist-
ance with development of pulmonary arterial hyper-
tension (PAH).67 When pulmonary vascular resistance
approaches and exceeds systemic vascular resistance,
ductal shunting reverses and becomes right to left, clas-
sically described as Eisenmenger syndrome. Clubbing
of toenails and spared (or mild) clubbing in the fingers is
pathognomonic for Eisenmenger syndrome in the set-
ting of a PDA.68 In this setting, patients may develop right
ventricular systolic failure, the most common cause of
death among patients with Eisenmenger syndrome.69
Infective Endocarditis
Historical estimates suggest an incidence of infective
endocarditis (IE) of ≈1% among patients with a PDA.70,71
Sadiq et al observed that, among 2908 children aged
<16 years admitted to a single pediatric cardiology
center over a 6- year time frame, 96 (3.3%) fulfilled di-
agnostic criteria for IE; PDA was the cardiac lesion in 14
children with IE (14.6%).72 Alternatively, of nearly 3 mil-
lion deaths in Sweden (1960– 1993), Thilen and Astrom-
Olsson reported 2 cases of IE as a complication of
PDA.73 Historically, risk of IE was commonly cited as an
indication for ductal closure beyond infancy74; however,
recent guidelines from the American Heart Association
no longer support use of antibiotic prophylaxis (outside
the first 6 months following definitive ductal closure)
among patients with PDA to prevent IE.75
DIAGNOSTIC ASSESSMENT OF THE
PDA
Diagnostic assessments vary according to the hemo-
dynamic significance of the ductus.
Unique Considerations in the Diagnosis of
PDA Among Preterm Infants
In contemporary medical settings, transthoracic echo-
cardiography is the preferred noninvasive modality to
assess ductal significance in preterm infants (Ta b l e 4).
Interrogation of ductal flow using 2- dimensional and color
flow Doppler can determine ductal size and shunt direc-
tion (Figure4A), as well as shunt volume (Figure4B).76,77
ASCERTAINMENT OF HEMODYNAMIC
SIGNIFICANCE
To target infants with the highest probability of de-
riving benefits following ductal treatment (described
below), health care providers have attempted to define
subgroups of preterm infants, based on clinical or so-
nographic criteria, with HSPDAs. However, lack of a
standardized or validated definition of HSPDA has led
to medical uncertainty about which infants with a diag-
nosis of PDA should receive treatment. This may relate,
at least in part, to the lack of consistency in the defini-
tions of HSPDA used to date in published randomized
clinical trials. A systematic review of the definition of
HSPDA used in published trials highlighted marked
variance with the use of arbitrary echocardiographic
thresholds, which were not validated against relevant
clinical outcomes.78
Historical Determinants of Ductal
Significance: An Oversimplification
The measurable physiological impacts of PDAs in
preterm infants are related to shunt volumes, intrin-
sic cardiopulmonary adaptive mechanisms, and du-
ration of exposure; therefore, arbitrary subjective or
single- point estimates of PDA sizes are unlikely to de-
fine hemodynamic significance accurately. However,
many trials relied solely on PDA diameters to adju-
dicate hemodynamic significance, which represents
an evaluative and physiological oversimplification.
This is attributable to the high likelihood of measure-
ment error related to geometric assumptions (circu-
lar in cross- section) or operator- dependent factors in
echocardiographic- based measurements of ductal
size.45,79,80,81 A PDA diameter of ≥1.5 mm is often used
to define hemodynamic significance, but the data
to support this practice are not evidence based.82
Moreover, several studies have demonstrated the
poor reliability of individual imaging measures and
the weak relationship of ductal diameter to echocar-
diographic indices of pulmonary or systemic blood
fl ow.83- 85 Although left atrial/aortic ratios are also
commonly used to determine ductal significance,
the metric is prone to significant operator- dependent
error and may be falsely normal in the context of a
large interatrial left- to- right shunt decompressing the
left atrium.84 In a comparative evaluation using mag-
netic resonance imaging, holodiastolic flow reversal
in the postductal arch was the most accurate and
reliable echocardiographic estimate of PDA shunt
volume.86
A Comprehensive Approach to Defining
HSPDAs
Rather than a unidimensional approach (eg, ductal
diameter), contemporary definitions of HSPDA in-
tegrate a comprehensive echocardiographic as-
sessment (Table5). The evaluative goal is to identify
preterm infants in whom ductal shunt volumes are
estimated to be primary pathological contributors
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 10
Backes et al PDA: A Contemporary Perspective
to current physiologic instability, considering other
concurrent pathologies (eg, lung immaturity and/
or ventilator- associated injury). The process of ad-
judication of hemodynamic significance should
incorporate clinical and echocardiographic param-
eters in a manner that helps health care provid-
ers recognize a hierarchal model of shunt volume
(small or large), rather than mere ductal patency.
The McNamara PDA staging system proposed
adjudicating hemodynamic significance based on a
composite of clinical and echocardiographic crite-
ria (see Table2, above).35 Prolonged exposures to
higher- volume shunts are associated with greater
risks of prolonged oxygen dependency87 or the
composite outcome of death or BPD.88 The PDA
scoring system recently proposed by El- Khuffash
et al, derived from a prospective evaluation of an
untreated cohort, combined gestational age with 4
Table 4. Diagnostic Assessments
Preterm infant*Diagnostic modalities*Term infant/adult
Continuous, “machinery” murmur diastolic rumble;
prominent LV impulse; tachycardia, tachypnea,
abdominal distention; prediastolic and postdiastolic
hypotension;
postductal systolic hypotension
Physical examinationDif ferential cyanosis (clubbing of the toe s, not the
fingers)§; high- frequency, diastolic decrescendo
murmur; holosystolic; peripheral edema
Cardiom egaly with LA an d LV enlargeme nt,
pulmonary artery dilation, and increased pulmonary
vascular markings
Chest radiographCalcif ications on the ductus; clear l ung fields; dilated
pulmonary arteries without significant cardiomegaly
Findings are highly variable and lack sensitivity/
specificity
ECGVentricular hyper trophy, and ST- segme nt or T- wave
depression
Diastolic flow reversal in postductal a rch; LA dilation
(LA:Ao); ductal diameter indexe d to LPA diameter;
PDA Vmax (CW); ductal left- to - right dia stolic fl ow; LVO;
pulmonary vein diastolic (PVD) Vmax; IVRT
Echocardiogram Right- to- left ductal shunting in systole can b e diff icult
to separate f rom adjac ent LPA; transe sophag eal
echocardiogram may be useful
CT Faster imaging and breath holding to identify
thromboembolic disease; cardiac gated CT can
identif y calc ifications; may reveal PDA not seen o n
echocardiogram
Magnetic resonance imaging Gold standard for noninvasive flow, function,
quantification, and anatomic evaluation without
ionizin g radiation for adults; may reveal PDA not seen
on echocardiogram
Safety and feasib ility of d evice closure dem onstrated
in infants weighing 700 g; venous- on ly approach
increasingly adopted
Cardiac catheterization Test occlusion to asse ss tolerance to clos ure (chang e
in PAP, systemic BP); pulmonary vascular reactivity
testing ca n identify resp onse to vasodilators
Lower values (cerebral, renal ) may be markers of
ductal significance; may be beneficial for longitudinal
assessment
Near- infrared spectroscopy Unclea r diagnostic value i n PDA assessment, but u sed
in other cardiovascular conditions and perioperative
settings
Unclear diagnostic value; insufficient evidence to
support universal monitoring
Biomarkers Unclear diagnostic value in PDA assessm ent, but used
in other cardiovascular conditions and perioperative
settings
… indicate s gold sta ndard for diagnosing PDA in preter m infants is transtho racic ec hocardiography; BP, blood pressure; CT, computed tomography; CW,
continuous wave; IVRT, isovolumetric relaxation time; LA, left atrial; LA:Ao, left atrial dia meter to aor tic root diameter rati o; LPA, left pulmonar y artery; LV, left
ventricular; LVO, LV output; PAP, pulmonary ar teria l pressure; PDA, patent du ctus ar teriosu s; and Vmax, maximum velocity.
*Findings described for preterm infants with hemodynamically significant PDA.
Findings described for adult patients with PDA and associated pulmonary arterial hypertension.
Findings are highly variable and lack sensitivity/specificity.
§Recomm end measurement of oxygen s aturatio n in upper a nd lower ex tremiti es in adults to assess for the pres ence of right- to- left shunting (dif ferenti al
cyanosis).
Figure 4. Ductal size and shunt direc tion evaluation (A ) and ductal shunt volume evaluation (B).
A, Echocardiographic verification of patent ductus ar teriosus (PDA) presence, with PDA size and shunt direction assessment. a,
Color flow of PDA . b, Pulsatile pattern with left- to- right low- velocity flow with wide differential between systole and diastole.
c, Restrictive pattern with higher velocity in both systole and diastole. d, Bidirectional pattern with right- to- lef t ductal flow during
systole. B, Echocardiographic examination of left atrial and left ventricular enlargement and quantification of ductal impact on cardiac
performance. a, Dilated left atrium (A) and ventricle (V). b, M- mode measurement, demonstrating a dilated left atrium indexed to the
aortic diameter. c, Elevated and pulsatile pulmonary venous flow, demonstrating high pulmonary venous diastolic flow. d, Shortened
isovolumetric relaxation time. e, Transmitral flow, demonstrating an early/atrial flow ratio of 1. f, Reversal of diastolic flow in the
postductal descending thoracic aorta. IVRT indicates isovolumetric relaxation time; LA:Ao, left atrial diameter to aortic root diameter
ratio; MPA, main pulmonar y artery; MV E/A, mitral valve early/atrial flow ratio; and PV D, pulmonary vein diastolic velocity.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 11
Backes et al PDA: A Contemporary Perspective
echocardiographic characteristics derived on post-
natal day 2, which provided accurate predictions of
the composite outcome of death or BPD (area under
the curve, 0.92 [95% CI, 0.86– 0.97]).89 However,
benefit from ductal closure based on these criteria
has not yet been demonstrated.
A
B
ac e
bd f
ab
cd
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 12
Backes et al PDA: A Contemporary Perspective
UNIQUE CONSIDERATIONS IN THE
DIAGNOSIS OF PDA AMONG OLDER
PATIENTS (TERM INFANT THROUGH
ADULTHOOD)
Transthoracic echocardiography provides adequate
imaging in term infants, but poor acoustic win-
dows in older patient populations may limit utility.
Transesophageal echocardiography can be used
with views in the upper esophagus using a clockwise
rotation from the aortic views.90 However, color flow
Doppler assessment can be misleading, particularly
in the presence of elevated pulmonary vascular resist-
ance, wherein right- to- left shunting in systole at the
ductus can be difficult to separate from adjacent struc-
tures, including the left pulmonary artery. These obser-
vations have led health care professionals to consider
diagnostic modalities for PDA in older patients beyond
echocardiography, including magnetic resonance im-
aging or computed tomography.
Among adolescent and adult patients with PAH,
cardiac magnetic resonance imaging or computed to-
mographic imaging may illuminate a PDA that was not
revealed during echocardiography. Cardiac magnetic
resonance imaging has long been considered the gold
standard for noninvasive flow assessments, function
quantification, and anatomic evaluation, providing useful
information on ductal morphology and characteristics,
without ionizing radiation. Alternatively, computed tomog-
raphy provides faster imaging and allows breath holding,
preferable for identifying in situ thromboembolic disease
that is prevalent in older patients with long- standing PDA
and associated PAH.91 In addition, cardiac- gated multi-
detector computed tomography scans can identify and
track the extent of calcifications found in subsets of adults
with PDA. This is particularly important to identify when
considering definitive closure (discussed below).92
Table 5. Comprehensive Echocardiographic Assessment of Ductal Significance in Preterm Infants
Echocardiographic
measures Significant PDA Limitations Measurement technique Normal range
Size of ductus
Indexed to LPA diameter PD A/ L PA >1.0 LPA dilation, high- volume
shunt
High- PS vi ew in 2D
Systemic blood flow (postductal aorta, celiac, MCA)
Flow pat terns: an tegrade,
absent, retrograde
diastolic flow
Flow reversal for dia stolic
steal; abnormal end organ
flow pattern
Accuracy determined by
measurement location angle
of insonation
Aorta: high- PS, PW Doppler
parallel to angle of f low
(diaphragm)
Celiac: sagittal view,
midabdominal
MCA: axi al view, tempo ral
fossa
Forward diastolic flow
Markers of pulmonary overcirculation
1. LVO LVO [>300] sec ondar y to
increased preload
Dependent on LVOT
morphology; minimize angle
correction
LV- VTI and Aorta: PW at
hinge points of aor tic valve
150– 300 mL/min per kg
2. Mitral valve E/A ratio E/A ≥1 indicates LA
pressure
Both unrel iable in the
setting of severe mitral valve
regurgitation
Apical 4Ch; PW Doppler
perpendicula r to the MV; at
tips of the le aflets
<1
3. IVRT IVR T <40 Apical 5Ch; PW at
intersection of inf low and
outflow on color Doppler
40– 60 ms
Left he art volume loadi ng
1. L A:Ao ≥1.6 Abnormality could be
secondary to LV dysfunction
PS, long- ax is M- mode
through aortic valve annulus
(LA:Ao)
LA: Ao <1.6
2. PV D- wave velocity PV diastol ic wave
velocit y [>0.5 m/s]
indicates PV return
Peak velo city de crease s as
PV dilates w ith larger shunts
Apical 4Ch PW Doppler
parallel to the RUPV inflow
0.2– 0.50 m/s
3. LPA end- diastolic
velocity
PBF leads to mean
and end- diastolic flow
velocity
LPA dilation commonly
occurs concurrently with
volume shunt
High- PS pa rallel to f low in
the LPA, dista l to the ducta l
insertion
<0.2 m/s
2D indicate s 2 dimens ions; 4Ch, 4 ch amber; 5C h, 5 chambe r; E/A, ratio of peak velocity bl ood flow f rom left ventricu lar rela xation in early di astole (E ) to peak
velocit y flow in late diastole caused by atrial contraction ( A); IVRT, isovolumetric rela xation time; LA, left atri al; LA:Ao, left atrial diameter to aortic root diameter
ratio; LPA, lef t pulmon ary ar tery; LV, left ventricular; LVO, LV outflow; LVOT, LVO tr act; MCA, middle cerebral ar tery; MV, mitral valve; PBF, pulmonary blo od flow;
PDA, patent du ctus ar teriosu s; PS, parasternal (vi ew); PV, pulmonar y vein; PW, pulse wave; and RUPV, right u pper PV; V TI, velocity time integral.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 13
Backes et al PDA: A Contemporary Perspective
In the setting of long- term ductal exposure, he-
modynamic assessment by cardiac catheterization
provides critical information on the risk/benefit profile
of ductal closure (Figure5). A 2018 joint statement
from the American Heart Association and American
College of Cardiology recommended, on the basis
of consensus expert opinion, that cardiac catheter-
ization can be useful in adult patients with PDA and
suspected PAH.36 In the setting of elevated pulmo-
nary vascular resistance, the PDA may serve as a
physiologic “pop- off” for the right ventricle, allowing
for egress of blood, albeit deoxygenated blood, from
the right ventricle to the systemic circulation. In this
setting, closure of the PDA may precipitate right ven-
tricular failure. Test occlusion of the ductus during
the catheterization can evaluate physiologic tolerance
of the right ventricle to shunt closure.93 In addition,
in the setting of concerns for PAH, performance of
pulmonary vascular reactivity testing during cardiac
catheterization can identify responsiveness to pulmo-
nary vasodilators.94
PDA TREATMENTS IN PRETERM
INFANTS
Indomethacin
Indomethacin, a cyclooxygenase inhibitor (see above)
targeting prostaglandin synthesis, is the prototypical
NSAID and is the most extensively studied medical
treatment for ductal closure among preterm infants.
Thirty- nine randomized trials (23 as prophylaxis,52 4 for
early treatment of an asymptomatic PDA,95 and 12 for
treatment of a symptomatic PDA96) demonstrate con-
sistent efficacy for achievement of ductal closure (RR
for persistent patency, 0.39 [95% CI, 0.35– 0.44], 0.41
[95% CI, 0.26– 0.65], and 0.40 [95% CI, 0.32– 0.50]),
at mean ages at treatment of <1, 2.5, and 6 days, re-
spectively. Despite efficacy in closing the ductus, ran-
domized trials have not identified benefits with respect
to most other outcomes (including mortality, BPD,
necrotizing enterocolitis, and neurodevelopment).40- 42
Indomethacin treatment is often followed by oliguria
and increases in serum creatinine levels, which have
Figure 5. American Heart Association guideline for management of noninfant patients with hemodynamically significant
patent ductus arteriosus (HSPDA).
Flowchart and guideline for the management of older patients and adults with HSPDA. Adapted from and based on recommendations
in Stout et al36. ECHO indicates echocardiography; IE, infective endocarditis; PASP, pulmonary arterial systolic pressure; PH,
pulmonary hypertension; and PVR, pulmonary vascular resistance.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 14
Backes et al PDA: A Contemporary Perspective
been linked to increased risks of spontaneous intesti-
nal perforation, particularly in infants also exposed to
postnatal corticosteroids.97,9 8
Ibuprofen
Ibuprofen has been compared with placebo or non-
treatment in 15 randomized controlled trials (6 as
prophylaxis99 and 9 as treatment64). Collectively, these
trials demonstrated efficacy for PDA closure compara-
ble to that of indomethacin (RR, 0.45 [95% CI, 0.40–
0.51]). Oral dosing appears to be more effective than
intravenous dosing (RR, 0.38 [95% CI, 0.26– 0.56]).64
Direct comparisons to indomethacin also support
comparable efficacy64,99 and suggest that oliguria and
necrotizing enterocolitis are less likely in infants treated
with ibuprofen.64 Other outcomes, with the exception
of ductal patency (and “rescue” NSAID treatment or
ligation), are not changed by ibuprofen prophylaxis99
or treatment.64 Comparable efficacy for ductal closure
and the lower risk of adverse effects have led some re-
viewers to designate ibuprofen as the NSAID of choice
for medical PDA closure.64
Acetaminophen
Recent reports have suggested that acetaminophen
(paracetamol) might be a less toxic, but similarly effec-
tive, alternative to indomethacin or ibuprofen for induc-
ing ductal closure. Most reports are anecdotal, without
controls, and there are few randomized trials.100 The 2
randomized trials comparing acetaminophen with pla-
cebo101 or no treatment102 enrolled a combined total
of 80 subjects and found that treatment reduced the
RR of ductal patency after 4 to 5 days of treatment
by 51% (RD, −0.21; RR, 0.49 [95% CI, 0.24– 1.00]).10 0
No differences in mortality, oxygen use at 36 weeks’
postmenstrual age, or other outcomes were observed.
Several small trials comparing acetaminophen with
ibuprofen or indomethacin found no differences in
rates of ductal closure between the treatments,100 sug-
gesting therapeutic equivalence. However, prospec-
tive nonrandomized data from the Early Treatment
Versus Delayed Conservative Treatment of the Patent
Ductus Arteriosus (PDA- TOLERATE) trial indicate that
acetaminophen is no more effective than conservative
treatment and is less effective than indomethacin in in-
ducing ductal closure.
103
Pharmacogenetics of Drug Treatment for
PDA
Recent evidence has shown that unpredictable re-
sponses to pharmacological PDA treatments (eg, in-
domethacin and ibuprofen) may reflect differences in
developmental trajectory (ontogeny), genetic variability
of drug- metabolizing enzymes, and drug targets.22 For
example, current weight- based dosing of indometha-
cin leads to variable drug exposures, with up to 14- fold
variation in drug concentrations 24 hours after identi-
cal intravenous drug dosing.104 To that end, genetic
variability in indomethacin metabolism may explain the
variability observed in drug exposures. Indomethacin
is metabolized by the cytochrome P450 enzymes, pri-
marily CYP2C9,105 and uridine 5’- diphospho- glucuron
osyltransferase (UGT) enzymes.10 6 Infants have low ex-
pression levels of CYP2C9 in fetal and early neonatal
life, and UGT enzymes are also expressed at relatively
low levels.107,108 For both enzymes, expression matures
rapidly during infancy.
In addition, CYP2C9 genotype has been associ-
ated with response to indomethacin in preterm infants
with PDA.109 The G allele of rs2153 6 28 was associated
with increased odds of response to indomethacin in
the case- control analysis (OR, 1.918 [95% CI, 1.056–
3.483]). Moreover, the polymorphism CYP2C92 was
associated with drug failure in a multicenter cohort
study of preterm infants receiving indomethacin for
PDA.110 The mechanisms by which variants in these
and other genes alter NSAID responses have not been
established, but may relate to the complex molecular
network regulating ductal patency before and after
birth.
Surgical Closure
Surgical ligation of the ductus, usually performed by
application of a surgical clip via a left posterolateral
thoracotomy,111 has the advantage of virtually univer-
sal achievement of ductal closure (although rare cases
of ligation of the left pulmonary artery or mainstem
bronchus have been reported). Only 4 procedural
PDA- closure trials have been conducted in preterm
infants.65 ,112 ,113 ,114 ,115 ,116 All trials evaluated open surgi-
cal ligation, rather than minimally invasive transcatheter
closure (discussed below), and were conducted before
the 1980s, before modern intensive neonatal care ad-
vances (eg, antenatal corticosteroids and surfactant)
that have allowed many preterm infants with PDAs to
survive.117 Prophylactic ligation of the ductus on the
day of birth for infants <1000 g in weight was associ-
ated with lower rates of necrotizing enterocolitis, more
frequent oxygen use and mechanical ventilation at
36 weeks, and no difference in mortality.65 Despite the
clear differences in ductal patency, ligation for symp-
tomatic PDA did not demonstrably decrease any ad-
verse outcome.40 The landmark National Collaborative
Study, comparing ligation with indomethacin for treat-
ment of persistent PDA, found that ligation was more
likely to produce ductal closure (RR, 0.04 [95% CI,
0.010.27]), but was associated with higher rates of
pneumothorax and retinopathy of prematurity, with-
out affecting mortality, chronic lung disease, or other
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 15
Backes et al PDA: A Contemporary Perspective
outcomes (by study design, the ductus was closed in
all subjects before study completion).
113
Ductal ligation is often followed within hours by car-
diorespiratory deterioration (postligation syndrome),
apparently attributable to altered afterload, leading to
impaired left ventricular systolic performance118; this
complication is more likely in infants who undergo liga-
tion at <30 postnatal days.119 Surgical ligation has also
been associated with increased risks of BPD114 ,12 0 and
neurodevelopmental impairment.
121 Long- term compli-
cations of surgical ligation include left vocal cord pa-
resis122 and scoliosis.123 In view of these observations,
rates of surgical ligation across US hospitals have de-
creased markedly over the past decade (8.4% in 2006
to 1.9% in 2015).55
Transcatheter Closure
Transcatheter closure is the procedure of choice for
definitive PDA occlusion in adults, children, and infants
≥6 kg, but application in smaller, preterm infants is a
more recent development.124 Availability of devices
specifically designed to address the unique ductal
morphology of preterm infants, including several with
the type F (fetal) PDA (see Figure2), coupled with in-
creasing experience among interventional teams,
has led to growing interest and use of this approach
(Figure6A). A multicenter, nonrandomized, single- arm
trial performed under the auspices of a US Food and
Drug Administration investigational device exemption
protocol and continued access program evaluated
safety and effectiveness of the Amplatzer Piccolo
Occluder device in 200 patients, including 100 weigh-
ing ≤2 kg and 33 weighing ≤1 kg. The trial demon-
strated high implant success rates (191/200 [95.5%]
and 99/100 [99%] at ≤2 kg) and low major complica-
tion rates (4/194 [2.1%]), with effective ductal closure
documented in 172 of 173 (99.4%) at 6 months. Five
infants were observed to have new evidence of mod-
erate tricuspid regurgitation on echocardiography fol-
lowing transcatheter closure, likely related to catheter
manipulation across the tricuspid valve.
126 Following
review of these results, the device was approved by
the US Food and Drug Administration for transcatheter
PDA closure in infants who weighed ≥700 g and were
aged ≥3 postnatal days.127
Evidence that the risk of an adverse event following
transcatheter PDA closure is inversely related to post-
natal age or procedural weight remains unclear.124,128
However, heterogeneity across studies in definitions
for complications, timing (intraprocedural or post-
procedural) of event reporting, and adjudication of
procedural- related adverse events limits the interpreta-
tion of available data on the risks of complications fol-
lowing transcatheter PDA closure. Recent procedural
modifications, including adoption of vascular access
using femoral venous, rather than femoral arterial, ap-
proaches, have led to marked reductions in the inci-
dence of postprocedure limb ischemia (Figure6B).129
Moreover, consensus- based guidelines outlining con-
temporary strategies to prevent and manage compli-
cations associated with transcatheter closure will likely
contribute to improved safety profiles.130
In the absence of direct comparisons, the optimal
treatment to achieve definitive ductal closure among
preterm infants remains unknown. Despite promising
short- term data, longer- term outcomes following tran-
scatheter PDA closure are lacking. Whether the greater
certainty of achieving ductal closure with this approach
will be associated with increased benefits to treated
infants (compared with NSAID treatment or conserva-
tive management, described below) also remains to be
determined.
124,129
Conservative Management
Despite evidence that a persistent ductus is associated
with worse outcomes, including death, randomized
trials of both pharmacological and surgical ligation
treatments to close persistent PDAs in preterm infants
have not demonstrated long- term benefits.40,41,43 To
that end, trials of treatment to close the ductus have
provided evidence that those measures, at least when
applied nonselectively, do not improve outcomes.40
These observations have led to increasing adoption
of conservative approaches to PDA management.
In conservative management, health care providers
avoid definitive (transcatheter or surgical) closure, while
awaiting the possibility of spontaneous closure.
131
With conservative management, several strategies
to manage consequences of the ductus are often used,
including fluid restriction, diuretics, systemic afterload
reduction, increases in positive airway pressures, or
maintenance of higher hematocrits; however, these
approaches have not been evaluated in systematic
randomized trials.41,131 Although data are mixed,
132,133
adoption of conservative management has not been
found to be associated with differences in outcomes.131
However, among “high- risk” subgroups, such as in-
fants born at <26 weeks’ gestation, with evidence of
HSPDAs producing demonstrable circulatory compro-
mise, or with persistent patency well beyond the ex-
pected age of spontaneous closure, questions on the
safety and effectiveness of conservative management
versus alternative treatments (definitive closure) remain
unanswered.3 ,13 2 ,13 3
Timing of Therapy
Because a speculative analysis of experiences with oral
or rectal indomethacin from the 1970s suggested that
treatment after 12.5 days of age may be less effective
than treatment at an earlier age,134 considerations of
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 16
Backes et al PDA: A Contemporary Perspective
treatment approaches for PDA have been influenced
by concerns that deferral of early treatment might result
in missed opportunities for effective NSAID treatment.
However, that analysis did not account for the expected
frequency of early spontaneous closure, which is com-
mon.3 Data from randomized clinical trials, including
those of indomethacin, do not support declining efficacy
with advancing postnatal age; trials that randomized
A
B
a
b
c
d
e
f
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 17
Backes et al PDA: A Contemporary Perspective
subjects at a mean postnatal age >7 days (range, 7.4–
20.1 days) demonstrated PDA- closure efficacy (RR, 0.35
[95% CI, 0.25– 0.48]) equivalent to trials of treatment at
earlier ages. In the PDA- TOLERATE trial, NSAID treat-
ment was followed by PDA closure in 46% of subjects
in the early (8±2 days) treatment group and in 44% of
those in the conservative treatment group who received
rescue treatment (at 21±8 days of age).80 ,10 3 The age at
which medical therapy becomes ineffective therefore re-
mains uncertain, but deferral of treatment into the fourth
postnatal week does not appear to compromise its utility
for achievement of ductal closure.
Postdischarge Treatment
At present, data on the posthospital outcomes of pre-
term infants who are discharged home with a persistent
PDA are lacking. Herrman et al reported that, among
21 infants discharged with an open ductus, 86% (18/21)
underwent spontaneous closure.135 This is consistent
with a recent report of spontaneous closure in 52 of 68
(76%) infants discharged with an open PDA, with contin-
ued evidence of closure beyond 2 years postnatal.136 In
a recent prospective multicenter study of 201 premature
infants discharged home with a PDA and followed up at
6- month intervals through 18 months of age, the authors
observed spontaneous ductal closure occurred in 47%
and 58% of infants at 12 and 18 months, respectively.13 7
In the absence of data, optimal outpatient surveillance
among preterm infants discharged with a persistent PDA
remains unknown.13 5,136
PDA TREATMENTS IN OLDER
PATIENTS (TERM INFANT THROUGH
ADULTHOOD)
Beyond the first postnatal months, term infants are
outside the window when pharmacological therapy
(indomethacin and ibuprofen) to close the ductus is
effective. Although diuretics may be used to treat pul-
monary overcirculation among term infants with a PDA
in the first months of life, decisions about the need for
ductal closure are largely driven by presence (or ab-
sence) of HSPDA, with consideration to the direction of
ductal shunting and pulmonary artery systemic pres-
sure and/or pulmonary vascular resistance indexed to
systemic pressure (see Figure5). The frequency and
timing of outpatient follow- up are determined on the
basis of PDA classification (Ta b l e 6).
Management of HSPDA
Transcatheter PDA closure remains the mainstay for
definitive ductal closure in older patients.
138 In patients
weighing >6 kg, most PDAs are amenable to tran-
scatheter occlusion, with the notable exception of the
type B ductus (see Figure2). According to 2011 guide-
lines from the American Heart Association on cardiac
catheterization in pediatric heart disease, based on
consensus expert opinion, transcatheter PDA occlu-
sion is indicated in older patients for the treatment of
an HSPDA with “left- to- right shunt that results in any of
the following: congestive heart failure, failure to thrive,
pulmonary overcirculation, or an enlarged left atrium or
left ventricle, provided the anatomy and patient size are
suitable” (class I recommendation, level of evidence
B).139
Management of PDA With Associated
PAH in Term Infants and Older Children
Among term infants and older children, high levels of
pulmonary artery pressure associated with unrestric-
tive ductal shunting may relate to excessive flow, and
not high pulmonary vascular resistance indicative of
pulmonary vascular disease. In this context, health
care providers may consider transcatheter closure fol-
lowing short- term pulmonary vasodilator testing (eg,
decrease of ≥20% in mean pulmonary artery pressure
Figure 6. Percuta neous patent ductus arteriosus (PDA) closur e (A) and intraprocedural imaging of percutaneous PDA
closure (B).
A, Illustration of percutaneous device release for closure of a PDA. An end hole catheter is used to cross the tricuspid valve into the right
ventricle (RV), then a soft, floppy- tipped wire is advanced across the PDA and into the descending aor ta (DAO) (data not shown). At this
point, the catheter in the right ventricle is removed over the wire, and a delivery catheter is advanced over the wire through the venous
sheath into the PDA and descending aor ta (data not shown). The device is advanced to the tip of the catheter (a). The device is deployed
under fluoroscopic and transthoracic echocardiography guidance within the PDA with careful attention to avoid device protrusion
into the aorta or pulmonary artery (b). When position is satisfactory, the device is released from the delivery cable (c). B, Radiographs
illustrating steps and final result of a percutaneous PDA closure procedure. a, Following femoral vein access, a 4F catheter is introduced
and advanced to the RV under fluoroscopic guidance, wherein a floppy- tipped wire is guided via this catheter through the PDA and into
the descending aor ta, after which a 4F delivery catheter is exchanged. b, An angiogram is obtained for configuration and dimensional
data of the PDA, which permits selection of the most appropriate device for closure. c, The device is then advanced through the deliver y
catheter and deployed, but not fully released. d, Additional echocardiographic imaging is obtained to confirm placement of the device.
e, Additional angiographic imaging to evaluate for aortic or left pulmonary obstruction attributable to the device. f, Device is released;
additional imaging to evaluate postrelease positioning and stability, residual shunting, and other clinical parameters (eg, presence of
new or increased tricuspid valve regurgitation) may be warranted. Reproduced from Barcroft etal125 with permission. Copyright ©2022
Elsevier. LPA indicates left pulmonary artery; and MPA, main pulmonar y artery.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 18
Backes et al PDA: A Contemporary Perspective
after administration of 100% oxygen and/or 80 parts
per million of inhaled NO) and a favorable hemody-
namic response to test occlusion.140 In a study by Niu
et al, the authors describe that younger patients with
pulmonary artery/systolic blood pressure ratios (car-
diac index) <0.8 during test occlusion were considered
to have favorable hemodynamics and underwent suc-
cessful PDA closure, despite many in the cohort hav-
ing met historical thresholds for being poor candidates
for intervention (pulmonary vascular resistance index,
>6 Wood units m2).94
Management of PDA With Associated
PAH in Older Patients and Adults
Recent American Heart Association/American
College of Cardiology guidelines emphasize that con-
siderations for ductal closure among older patients
be made in the context of evaluation of left- to- right
shunting and hemodynamic assessment for PAH (see
Figure5).36 Correspondingly, European guidelines
suggest pulmonary/systemic shunt ratios of <1.5 and
pulmonary vascular resistances of >5 Wood units as
prohibitive for ductal closure in adults.69 Supportive
pharmacologic treatments (eg, endothelin recep-
tor antagonists and phosphodiesterase- 5 inhibitors)
have been shown to improve functional capacities in
adult patients with Eisenmenger syndrome, includ-
ing marked increases in survival advantage for those
on PAH therapies versus not on PAH therapies (97%
versus 69%; P<0.01).141 Before transcatheter closure,
balloon test occlusion provides insight on risk/benefit
profiles, which are particularly valuable for patients
with evidence of PAH.
Definitive Closure (Transcatheter or
Surgical Ligation)
Transcatheter PDA closure in older patients remains
the mainstay for definitive ductal closure.
138 Following
hemodynamic assessment, for patients with unfa-
vorable ductal morphology for transcatheter closure,
surgical intervention remains feasible via both video-
assisted thoracoscopic or open thoracotomy, with high
degrees of success and low complication rates.14 2,143
Management of “Silent” or Small PDAs
Since the first surgical PDA ligation by Gross in 1939,
thresholds for intervention on the basis of IE risk have
been appropriately recalibrated as a function of the
advent of antibiotic treatment and the ability to offer
transcatheter PDA occlusion.
144 ,145 In other words, pre-
vious indications to close were based on prevention
of endarteritis, wherein closing the PDA would curtail
the need for long- term antibiotic prophylaxis. Because
“silent” or small PDAs are not considered HSPDA (see
Table2, above), societal guidelines no longer recom-
mend antibiotic prophylaxis; thus, the rationale for
closing “silent” or small PDAs no longer exists.36 ,75
CONCLUSIONS
PDA is a complex pathophysiology, resulting in mark-
edly variable clinical consequences. To provide more
targeted, individualized treatments, a better under-
standing of the molecular mechanisms of ductal
closure, the effect of each patient’s clinical and echo-
cardiographic biomarkers on both treatment success
and improved outcomes, as well as genetic variants
Table 6. Recommended Frequency of Outpatient Follow- Up and Treatment Among Term Infants, Older Children, and
Adults With PDA
Classification
Assessment intervals and treatment options
Pediatric/ACHD
cardiologist ECG TTE
Pulse
oximetry
Exercise
test*Treatment
“Silent ” or trivial PDA36– 60 months 36– 60
months
36– 60
months
As needed As needed None
Small PDA24 mon th s 24 mo nths 24 mo nths As needed As needed None
HSPDA§ or PDA with mild
or moderate PH
6– 12 mo nt h s 12 mo nt hs 12 mo nt h s Each visit 12– 24 mont hs Closure is recommended if PDA
causing LAE /LVE with net lef t-
to- right sh unt, PASP <50% of
systemic, a nd PVR <1/3 systemic
PDA with severe PH and/or
Eisenmenger syndrome
3– 6 months 12 m on th s 12 m o nt hs Each visit 6– 12 mo nt h s Closure is NOT reco mmende d with
net left- to- right shu nt and PASP or
PVR ≥2/3 sys temic
ACHD indic ates adult congenital hea rt dise ase; HSPDA, h emodynamically significant PDA; LAE, left atria l enlarg ement; LVE, lef t ventricular enl argeme nt;
PASP, pulmonary ar tery systolic pre ssure; PDA, p atent ductus arter iosus; PH, pu lmonar y hypertension; PVR, pulmonar y vascular resistance; and T TE,
transthoracic echocardiography.
*Six- minute walk test or cardiopulmonary exercise test.
No hemodynamic or anatomic consequences of the PDA.
Small shunt that is not HSPDA and is asymptomatic.
§HSPDA=LAE/LVE and/or su stained pulmona ry blo od flow to syste mic bloo d flow ratio (Qp/Qs) ≥1.5.
PDA closure c onside red: net lef t- to- right shu nt if PASP ≥50% systemic and/or PVR is ≥1/3 systemi c.
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 19
Backes et al PDA: A Contemporary Perspective
in drug metabolism and drug targets should be pri-
oritized.
146 Irrespective of patient age, incorporating
clinical and cardiac imaging parameters that recog-
nize a hierarchal model of adverse ductal sequalae is
recommended. The critical need for contemporary,
pragmatic clinical trials that evaluate the impact of ex-
isting PDA treatments on important patient outcomes
is acknowledged.
Affiliations
Center for Perinata l Research, The Abigail Wexn er Research Insti tute at
Nationwi de Children’s Hospital, Columbu s, OH (C.H.B., J.L.S.); Division
of Neonatology, Nationwide Child ren’s Hospita l, Columbus, OH (C.H.B.,
J.L.S.); Department of Pedi atrics, T he Ohio St ate Univer sity Co llege of
Medici ne, Columbus, OH (C.H.B., J.L.S., M.L.M., V.G.); The Heart Center,
Nationwi de Children’s Hospital, Columbu s, OH (C.H.B., M.L.M., V.G.); Duke
University Pediatric and Congen ital Hea rt Disease Center, Durham, NC
(K.D.H.); Duke Clini cal Research Institute, Durham, NC (K.D.H.); Department
of Pediatrics (E.L.S.), and Dep artme nt of Pharm acolog y (E.L.S.), Vanderbilt
University Medical Ce nter, Nashvill e, TN; ; Divis ion of Epid emiolo gy, College
of Public Health, The Ohio State U niversity, Columbus, OH (J.L.S.); Divisi on
of Neonatology (T.R.L.), and Division of Cl inical Pharmacology, Toxicology
and Therapeuti c Innovatio n (T.R.L.), Children’s Mercy Kansas Cit y, Kansas
City, MO; Depa rtmen t of Pediatrics, University of Missouri– Kansas Ci ty
School of M edicin e, Kansas C ity, MO (T.R.L.); Depar tment of Pae diatric s,
University of Toronto, Onta rio, Canada (D.E.W.); Depar tment of Newborn a nd
Developmental Paediatrics, Sunnybrook Health Science Center, Toronto,
Ontari o, Canada (D.E.W.) Division of Neonata l and Development al Medicine,
Depar tment of Pediatric s, Stanford Univers ity Sch ool of Medicine, Luc ille
Packard C hildren’s Hospita l, Stanford, CA (S.B., W.E.B.); Center for Integ rated
Brain Re search, University of Washington School of Medicine, Sea ttle,
WA (C.V.S.); Department of Pediatrics (P.J.M.), and Depar tment of Inte rnal
Medici ne (P.J.M.), University of Iowa, Iowa City, IA; Center for Cardiovascular
Research, The Abigail Wexner Resea rch Institute at Nationwide Children’s
Hospital, Columbus, OH (V.G.); and Department of Molecular Gene tics, The
Ohio State Univers ity, Columb us, OH (V.G.).
Sources of Funding
None.
Disclosures
Drs Backe s, Slaughter, and McNama ra are investigators o n the upcoming
“PIVOTAL” (Percuta neous Intervention Versus O bser vational Trial of Arterial
Ductus in Low- We ight Infa nts; UG3HL161338- 01), funded by the National
Heart, Lung, and Blood Insti tute. For the conduct of th is study, the
investigators received additional funding from Abbott to support the trial;
however, Abbott has no influence in the desig n, conduc t, or execution
thereof of the study. The remaining authors have no disclosures to report.
REFERENCES
1. Gentile R, Stevenson G, Dooley T, Franklin D, Kawabori I, Pearlman
A. Pulsed Doppler echocardiographic determination of time of ductal
closure in normal newborn infants. J Pediatr. 1981;98:443– 448. doi:
10.1016/s 0 022 - 3 476( 81)8 0719 - 6
2. Koch J, Hensley G, Roy L, Brown S, Ramac iotti C, Rosenfeld
CR. Prevalence of spo ntaneous closure of the ductu s arter io-
sus in neo nates at a bir th weigh t of 1000 grams or less. Pediatrics.
20 0 6 ;117:111 3 – 1121. doi: 10 .1542 /p ed s. 20 0 5- 152 8
3. Semberova J, Sirc J, Miletin J, Kuce ra J, Berka I, Sebkova S, O’Su llivan
S, Frankli n O, Stranak Z. Spontane ous clos ure of patent d uctus ar teri-
osus in inf ants ≤1500 g. Pediatrics. 2017;140:e20164258. doi: 10 .1 54 2 /
pe ds .20 16- 425 8
4. Sc hneide r DJ, Moore JW. Congenital hear t disease for the adult car-
diologist: patent ductus arteriosus. Circulation. 20 0 6;114:1873 – 18 82.
doi: 1 0.11 61 /C I R C U L AT I O N A H A .10 5 . 59 2 0 6 3
5. Clyman RI, Wal eh N, Black SM, Riemer RK, Mauray F, Chen YQ.
Regulation of ductus arter iosus patency by nitr ic oxide in fetal lamb s:
the role of ge station, ox ygen tension, and va sa vasor um. Pediatr Res.
1998;43:633– 644. doi: 10.1203/00006450- 199805000- 00012
6. Smith GC, Coleman RA, Mc Grath JC. Characterization of dilator pros-
tanoid re ceptors in the fetal ra bbit duc tus arte riosus. J Pharmaco l Exp
Ther. 1994;271:390 – 39 6.
7. Leonhardt A , Glaser A, Wegman n M, Schran z D, Seyber th H, Nusin g
R. Expression of p rostano id recepto rs in huma n ductus arterio sus. Br
J Pharmacol. 2003;138:655– 659. doi: 10.1038/sj.b jp. 0705092
8. Michela kis E, Rebeyka I, Bateson J, Olley P, Puttagun ta L, Arche r
S. Voltage- gated potassium channels in human ductus arteriosus.
Lancet. 200 0; 35 6:13 4– 13 7. doi: 10.1016/S 0140 - 6 736 (0 0) 024 52- 1
9. Shelton EL, Ector G, Gal indo CL, Ho oper CW, Brown N, W ilkerso n I,
Pfalt zgraff ER, Pari a BC, Cotton RB, Stolle r JZ, et al. Transcription al profiling
reveals ductus arteriosus- specific genes that regulate vascular tone. Physiol
Genomics. 2014;46:457– 466. doi: 10.1152/physiolgenomics.00171.2013
10. Waleh N, Reese J, Ka jino H, Roman C, Seidn er S, McCurnin D, Cly man
RI. Oxygen- induced tension in the shee p ductus a rteriosus: effects of
gestation on potassium and calcium channel regulation. Pediatr Res.
2009;65:285– 290. doi: 10.1203/PDR.0b013e31819746a1
11. G onzale z- Luis G, Ghiradello S, Bas- Sua rez P, Cavallaro G, Mosc a F,
Clyman R I, Villamor E. Platele t counts an d patent duc tus arte riosus
in preterm i nfants: an up dated syste matic revi ew and meta- analysis.
Front Pediatr. 2020;8:613766. doi: 10.338 9/fped. 2020.613766
12. Sallmon H, Webe r SC, Huning B, Stein A, H orn PA, Metze BC, Dame C,
Buhrer C, Felderhoff- Muser U, Hansmann G, et al. Thrombocytopenia
in the first 24 hours after birth and incid ence of pate nt ductus arteri o-
sus. Pediatrics. 2012;130:e623 – e63 0. doi: 10.1542/peds.2012- 0499
13. Kahvecioglu D, Erdeve O, Akduma n H, Ucar T, Alan S, Cakir U, Yildiz
D, Atasay B, Ars an S, Atalay S. Influence of platel et count, pl atelet
mass ind ex, and plate let function on the spontane ous clos ure of duc-
tus arteriosus in the prematurity. Pediatr Neonatol. 2018;59:53– 57.
doi: 10.1016/j.pedneo.2017.01.00 6
14. Toizumi M, Motomura H, Vo HM, Takahashi K, Pham E, Nguye n HA,
Le TH, Hash izume M, Ar iyoshi K, Dang DA, et al. Mortality associ-
ated with pulmonary hypertension in congenital rubella syndrome.
Pediatrics. 2014;134:e519– e 526. doi: 10.1542/peds.2013- 4184
15. O rofino DHG, Passos S RL, de Oliveira RVC, Fari as CVB, Leite M, Pone
SM, Pone M, Teixeira Mendes H AR, Moreira MEL, Nie lsen- Saines K.
Cardiac f indings in infants w ith in utero ex posure to Zi ka virus- a cross
sectional study. PLoS Negl Trop Dis. 2018;12:e00 063 62. do i: 10 .13 71 /
journal.pntd.0006362
16. Reece AS, Hulse GK. Cannabis teratolog y explai ns current patterns
of coloradan cong enital d efects: the c ontribu tion of increased can-
nabinoid exposure to rising teratological trends. Clin Pediatr (Phila).
2019;58:1085– 1123. doi: 10 .117 7/ 0 0 09 9 2 2 81 98 6 12 81
17. Reece AS, Hul se GK. Broad spectr um epide miological contributi on
of cannabis and othe r substa nces to the teratologic al profile of north-
ern New So uth Wales: ge ospatia l and caus al infere nce analysis. BMC
Pharmacol Toxicol. 202 0; 21:75 . d oi: 10.1186/s40360- 020- 00450- 1
18. Yarboro MT, Gopal SH, Su RL, Morgan TM, Reese J. Mouse mo dels
of patent duc tus arte riosus (PDA) and thei r relevance for huma n PDA.
Dev Dyn. 2022;251:424– 44 3. doi: 10.1002/dvdy.408
19. Pierpont ME, Brueckner M, Chung WK, Garg V, Lacro RV, McGuire
AL, Mital S, Priest JR, Pu WT, Roberts A, et al. Genetic basis for
congen ital hea rt disease: revis ited: a scie ntific st atement from the
American Heart Association. Circulation. 2018;138:e653 – e711. doi:
10.1161/CIR.0000000000000606
20. L avoie PM, Pham C, Jang KL. Heritab ility of bronchop ulmonary dys-
plasia, defined accordi ng to the consensus sta tement of the National
Institute s of Health. Pediatrics. 2008;122:479– 485. doi: 1 0.1 5 42 /
pe d s . 20 0 7- 2 313
21. Dagle JM, Lepp NT, Cooper ME, Schaa KL, Kelsey KJ, Orr KL, Caprau
D, Zimmerm an CR, Stef fen KM, Jo hnson KJ, et a l. Determi nation of
genetic p redisposition to pa tent ductu s arteriosus in preterm infa nts.
Pediatrics. 20 09;123:1116– 1123. doi: 10.1542/peds.2008- 0313
22. Lew is TR, She lton EL, Van Dri est SL, Kannanker il PJ, Reese J.
Genetic s of the patent ductus ar teriosus (PDA) and pharmac ogeneti cs
of PDA treatment. Semin Fetal Neonatal Med. 2018;23:232– 238. doi:
10.1016/ j.s in y.2018.0 2.0 06
23. Bokenk amp R, DeRuiter MC, van M unsteren C, Gittenb erger- de
Groot AC. Insights into the pathogenesis and genetic background
of patency of th e ductus arterio sus. Neonatology. 2010;98:6– 17. doi:
10.1159/000 262481
24. Mitche ll SC, Korones S B, Berend es HW. Congenital heart dis ease in
56,109 bir ths. Incidence an d natural h istory. Circulation. 1971;43:323–
332. doi: 10 .116 1/ 0 1. c i r. 43. 3 . 3 23
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 20
Backes et al PDA: A Contemporary Perspective
25. Connuck D, Sun JP, Super DM, K irchne r HL, Fradley LG, H arcar-
Sevcik RA, Salvator A, Singe r L, Mehta SK . Incide nce of patent ductus
arteriosus and patent foramen ovale in normal infants. Am J Cardiol.
200 2;8 9:24 4– 247. doi: 10.1016/s0002- 9149(01)02214- 7
26. Hundscheid T, van den Broek M, van de r Lee R, de Bo ode WP.
Understanding the pathobiology in patent ductus arteriosus in
prematurity- beyond prostaglandins and oxygen. Pediatr Res.
2019;86:28– 3 8. doi: 10.103 8/s 4139 0- 019- 0 387- 7
27. Friedma n WF. The intrinsic physiologic propertie s of the de-
velopin g hear t. Prog Cardi ovasc Dis. 19 72 ;1 5: 87– 111. do i:
10.1016/0033- 0620(72)90006- 0
28. Ne lin LD, Wearden ME, Welty SE, Hansen T N. The ef fect of blo od
flow and left atri al pressure on the DLCO in lambs and sheep. Respir
Physiol. 1992;8 8:333– 342. doi: 10.1016/0034- 5687(92)90007- j
29. Alpan G, Schee rer R, Bland R, Clyman R. Patent ductus ar teri-
osus increases lung fluid filtrati on in preter m lambs. Pe diatr Res.
1991;3 0 :6 16 6 21. doi: 10.1203/00006450- 199112000- 00026
30. Perez Fo ntan JJ, Cly man RI, Mau ray F, Heymann MA, Roman C.
Respiratory effects of a patent ductus arteriosus in premature new-
born lambs. J Appl Physiol (1985). 1987;63:2315– 2324. doi: 1 0.11 52 /
jappl.1987.63.6.2315
31. Ikegami M, Jacobs H, Jobe A. S urfac tant function in respirator y
distres s syndrom e. J Pediatr. 1983;102:443– 447. doi: 10.1016/
s0022- 3476(83)80673- 8
32. Matsuda M, Ande rson- M orris J, Ra j JU. Microvascular pressure s in
isolated p erfu sed immature lamb lungs: effects of flow rate, left atrial
pressure and sur factant therapy. Biol Neonate. 1996;70:349– 358. doi:
10.1159/000 244386
33. C lyman RI, M auray F, Heymann MA, Roman C. Ca rdiovasc ular ef fects
of patent duc tus arte riosus i n preterm la mbs with re spiratory distress.
J Pediatr. 19 8 7;111:5 7 9 5 8 7. doi: 10.1016/s0022- 3476(87)80126- 9
34. Keuster s L, Purna J, De shpand e P, Mertens L, Shah P, McNamara
PJ, Weisz DE, Jain A. C linica l validit y of systemi c arter ial steal a mong
extremely preter m infants with persistent patent ductus ar terio sus.
JPerinato l. 2021;41:84 – 92. doi: 10.1038/s 41372- 020 - 06 63 - 8
35. McNamara PJ, Sehgal A . Towards rationa l management of the p atent
ductus arteriosus: the need for disease staging. Arch Dis Child Fetal
Neonatal Ed. 2 007;92:F424– F427. doi: 10 .113 6 /a d c . 20 0 7.118 117
36. Stou t KK, Dani els CJ, Aboulhosn JA, Bozkur t B, Brobe rg CS,
Colman J M, Crumb SR , Dearani JA, Fulle r S, Gurv itz M, et al. 2018
AHA/ACC guideline for the management of adults with congenital
heart disease: executive su mmar y: a repor t of the American Co llege
of Cardiology/American Heart Association Task Force on Clinical
Practice Guidelines. Circulation. 2 019;139:e637– e 697. doi: 10 .11 61 /
CIR.0000000000000602
37. Kidokoro H, Ande rson PJ, Doyle LW, Woodward LJ, Neil JJ, Inder
TE. Brain injury a nd altered brain grow th in prete rm infants: predic-
tors and pro gnosis. Pediatrics. 2014;134:e444 – e 453. doi: 1 0.1 5 42 /
peds.2013- 2336
38. Mirza H, Garcia J, Mc Kinley G, Hubbard L, Sensing W, Sch neider
J, Oh W, Wadhawan R. Du ration of si gnificant patent ductus ar teri-
osus and bronchopulmonary dysplasia in extremely preterm infants.
JPerinato l. 2019;3 9:16 48 – 1655. doi: 10.103 8/s 41372- 019- 04 96 - 5
39. R yder RW, Shelton JD, Guinan ME. Necrotizing enterocolitis: a pro-
spective multicenter investigation. Am J Epi demiol. 1980;112:113– 123.
doi: 10.1093/oxfordjournals.aje.a112960
40. Benit z WE. Treatment of p ersistent patent ductus ar teriosus in preter m
infants: tim e to accept th e null hypothesis? J Perinatol. 2010;30:241–
252. doi: 10 .1038 /j p.2 010. 3
41. Benitz WE. Lear ning to live w ith patency of the ductus arteriosus in
preterm infants. J Perinatol. 2011;31(suppl 1):S42– S48. doi: 10 .1038 /
jp. 2 010 .175
42. Benitz WE; Ame rican Academy of Pediatric s Commit tee on Fetus
Newborn. Patent ductus arter iosus in p reterm infants. Pediatrics.
2016;137:e20153730. doi: 10 .1542 /p ed s. 20 15- 373 0
43. Noori S, McCoy M, Fr iedlic h P, Bright B, Gottip ati V, Seri I, Sekar K.
Failure of ductus arteriosus closure is associated with increased mor-
tality in preterm infants. Pediatrics. 20 09 ;123 :e13 8– e14 4. doi: 10 .15 42 /
peds.2008- 2418
44. Gournay V, Roze JC, Kuster A, Daoud P, Cambonie G, H ascoet JM,
Chamboux C, Blanc T, Fichtner C, Savagner C, et al. Prophyl actic
ibuprofen versus placebo in very premature infants: a randomised,
double- blind, placebo- controlled trial. Lancet. 2 00 4;3 64:19 39 – 19 44.
doi: 10.1016/S0140- 673 6( 04 )17476- X
45. K luckow M, Jeffer y M, Gill A, Evans N. A rand omised p lacebo -
controll ed trial of early tre atment of th e patent duc tus arte riosus.
Arch Dis C hild Fetal Neonatal Ed. 2014;99:F99– F104. doi: 10 .113 6 /
archdischild- 2013- 304695
46. Mi tra S, Scrivens A, von Ku rsell AM, Disher T. Early treatment ver-
sus expectant management of hemodynamically significant patent
ductus arteriosus for preterm infants. Cochrane D atabase Syst Rev.
2020;12:CD013278. doi: 10.1002/1465185 8.C D013278 .pu b2
47. Clyman RI, H ills NK, L iebowitz M, Johng S. Relationship bet ween
duration of infant exposure to a moderate- to- large patent ductus ar-
teriosus shunt and the risk of developing bronchopulmonary dyspla-
sia or death before 36 weeks. Am J Perinatol. 2020;37:216– 223. do i:
10.1055 /s- 0039- 1697 672
48. Beni tz WE, Choc k VY. Prolonge d ductal patency in preterm in-
fants: does it matter ? J Pediatr. 20 21;2 29 :12– 14.e11. doi: 10.1016/j.
jpeds. 2020.10.059
49. Roberts G, A nderson PJ, De Luca C, Doyle LW; Victorian Inf ant
Collaborative Study Group. Changes in neurodevelopmental outcome
at age eight in geogr aphic co horts of c hildren born at 22- 27 weeks’
gestational age during the 1990s. Arch Dis Child Fet al Neonat al Ed.
2010;95:F90– F94. doi: 10 .113 6 /a d c . 2 0 09 .1 65 4 8 0
50. Larroque B, Anc el PY, Marchand- Mar tin L, Cambonie G, Fres son J,
Pierrat V, Roze JC, Marpeau L, Thiriez G, A lberge C, et a l. Special
care and school difficulties in 8 - ye ar- old very preterm chi ldren: the
Epipage c ohort study. PLoS One. 2011;6:e21361. doi: 10.1371/journal.
pone.0021361
51. Mukerji A, Shah V, Shah PS. Periventricular/intraventricular hemor-
rhage and neurodevelopmental outcomes: a meta- analysis. Pediatrics.
2015;13 6:1132 – 1143. doi: 10.154 2/ pe ds .2 015 - 0 944
52. Fowlie PW, Davis PG, McGuire W. Prophylactic intravenous indometh-
acin for preventing mortality and morbidity in preterm infants. Cochrane
Database S yst Rev. 2010;2010:CD 000174. doi: 10.10 02 /14651858 .
CD000174.pub2
53. Ment L R, Dunca n CC, Ehrenk ranz RA , Klein man CS, Pit t BR, Taylor
KJ, Scott DT, Stewart W B, Gettne r P. Randomized indomethacin trial
for prevention of intrave ntricular hemo rrhage i n very low birth weight
infants. J Pediatr. 1985;107:937– 943.
54. Schmidt B, D avis P, Moddemann D, Ohlsson A, Rober ts RS, Saig al S,
Solimano A, Vinc er M, Wright LL; Trial of Indom ethacin Prophyla xis in
Preterms Investigators. Long- term effects of indomethacin prophylaxis
in extre mely- low- birth- weight infants. N Eng l J Med. 2001;344:1966
1972. doi: 10.1056/NEJM200106283442602
55. Bixler GM, Powers GC, Cla rk RH, Walke r MW, Tolia VN. Change s
in the diag nosis and management of paten t ductus arterio sus from
2006 to 2015 in United States neon atal inten sive care u nits. J Pediatr.
2017;189 :105– 112. doi: 10.1016/ j.j pe ds .2017.05.0 24
56. Rees e J, Shelton EL, S laughter JC, McNama ra PJ. Prophylactic in-
domethacin revisited. J Pediatr. 2 017 ;18 6 :11 – 1 4. e11 . doi: 10.1016/j .
jpeds.2017.03.036
57. DeMauro SB, Schmi dt B, Roberts RS. Why would a sane clinici an not
prescribe prophylactic indomethacin? Acta Paediatr. 2011;100:636.
doi: 10.1111 / j.1 6 51- 2 2 27. 2 0 11. 022 16 . x
58. Foglia EE, Roberts RS, Stoller JZ, D avis PG, Ha slam R, Schmidt B;
Trial of Indomethacin Prophylaxis in Preterms I. Effect of prophylac-
tic indom ethacin in extreme ly low bir th weight infants ba sed on the
predicted risk of severe intraventricular hemorrhage. Neonatology.
2018;113:183 – 186 . doi: 10.1159/000485172
59. Jensen EA, Foglia EE, Schmidt B. Association between prophylac-
tic indomethacin and death or bronchopulmonary dysplasia: a sys-
tematic rev iew and met a- analy sis of obse rvatio nal studi es. Semi n
Perinatol. 2018;42:228– 234. doi: 10.1053/j.semperi.2018.05.005
60. Schmidt B, Se shia M, Sha nkaran S, Mildenhall L, Tyson J, Lui K, Fok
T, Roberts R; Trial of Indomethacin Prophylaxis in Preterms I. Ef fects of
prophylactic indomethacin in extremely low- birth- weight infants with and
without adequate exposure to antenatal corticosteroids. Arch Pediatr
Adolesc Med. 2011;165:642– 646. doi: 10.1001/archpediatrics.2011.95
61. Weisz DE, More K, McNa mara PJ, Shah PS. PDA ligation and health
outcome s: a meta- analysis. Pediatrics. 2014;133: e102 4– e10 4 6. doi:
10.1542/peds.2013- 3431
62. Weisz DE, M irea L, Rosenberg E, Ja ng M, Ly L, Church P T, Kelly E,
Kim SJ, Jai n A, McNamara PJ, et al. Asso ciation of p atent ductus ar-
teriosu s ligation with death or neurod evelopmental impairment among
extremely preter m infants. JAMA Pediatr. 2017;17 1:4 4 3 44 9 . doi:
10.1001/jamapediatrics.2016.5143
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 21
Backes et al PDA: A Contemporary Perspective
63. Hsu KH, Nguyen J, Dekom S, Ra manathan R, Noori S. Effects of
patent duc tus arte riosus o n organ blood flow in i nfants born very
preterm: a prospective study with serial echocardiography. J Pediatr.
202 0;216 :95 – 10 0.e10 2. doi: 10.1016/j.jp ed s. 2019.08 .057
64. Ohlsso n A, Walia R, Shah SS . Ibuprofen for the tr eatment of patent d uc-
tus arte riosus in preterm or l ow birth weight (or both) infants. Cochrane
Database S yst Rev. 2020;2:CD003481. doi: 10.10 02 /14651858 .
CD003481.pub8
65. Cassady G, Crouse DT, Kirklin JW, Strange MJ, Joiner CH, G odoy G,
Odrezin GT, Cutter GR, Kirklin JK, Paci fico AD, et al. A randomi zed,
controll ed trial of very ea rly prophylactic l igation of the ductus a rteri-
osus in babies who we ighed 1000 g or less at birth. N Eng l J Med.
198 9; 32 0:1511– 1516.
66. Shaf fer ML, Bau d O, Lacaze - Masmonte il T, Peltoniemi OM, B onsante
F, Watterberg KL. Ef fect of prophylaxis for ear ly adren al insuf ficiency
using low- dose hydrocortisone in very preterm infants: a n individual
patient data meta- analysis. J Pediatr. 2019;2 07:136 – 142.e135. doi:
10.1016/ j.j pe ds .20 18.10.0 04
67. Arvanitaki A , Giannakoulas G, Baumgar tner H, Lammers A E.
Eisenmenger syndrome: diagnosis, prognosis and clinical manage-
ment. Heart. 202 0;10 6:16 38 – 16 45 . doi: 10.1136/heartjnl- 2020- 316665
68. Srinivas SK, Manjunath CN. Dif ferenti al clubbing and cya nosis: cla ssic
signs of patent ductus arteriosus with Eisenmenger syndrome. Mayo
Clin Proc. 2013;8 8:e10 5– e10 6 . doi: 10.1016/j.mayocp. 2013.02.016
69. Baumg artne r H, Bonhoeffer P, De Groot NM, de H aan F, Deanfie ld
JE, Galie N, G atzouli s MA, Gohlke- Baerwol f C, Kaemmerer H, Kiln er
P, et al. ESC guidelines for the manageme nt of grown- up congenital
heart disease (new version 2010). Eur Heart J. 2010;31:2915– 2957.
doi: 10.1093/eurheartj/ehq249
70. Campbell M. Natural his tory of persistent d uctus ar terios us. Br Heart
J. 1968 ;3 0:4 – 13. doi: 10 .113 6 / h r t . 3 0 .1. 4
71. Johnson DH, R osentha l A, Nadas AS. A fort y- ye ar review of bacterial
endocarditis in infancy and childhood. Circulation. 1975;51:581– 588.
doi: 10.11 61 /0 1. c i r. 51 .4 . 5 81
72. Sadiq M, Latif F, Ur- R ehman A . Analysis of infective enda rteritis in pat-
ent ductus arteriosus. Am J Cardiol. 2 00 4; 93 :513 515. doi: 10 .1016/j .
amjcard.20 03.10.062
73. Thilen U, Astrom- Olsson K . Does the risk of infe ctive endarter itis justify
routine patent ductus arteriosus closure? Eur Hear t J. 1997;18:503–
506. doi: 10.1093/oxfordjournals.eurheartj.a015272
74. Heuc han AM, Clyman RI. Ma naging th e patent duc tus arte rio-
sus: curre nt treatme nt options. Arch Dis C hild Fetal Neonatal Ed.
2014;99:F431– F436. doi: 10.1136/archdischild- 2014- 306176
75. Baddour LM, Wilson WR, Baye r AS, Fowler VG Jr, Tleyjeh IM, Ry bak
MJ, Barsic B, Lockhart PB, Gewitz MH, Levison ME, et al. Infective e n-
docarditis in adults: diagnosis, antimicrobial therapy, and management
of compli cations: a scientif ic statement for hea lthcare p rofessionals
from the Am erica n Hear t Associ ation. Circulation. 2015;132:14 35
148 6. doi: 10.1161/CIR.0000000000000296
76. Van Laere D, Van Overmeire B, Gupta S, El- Khuffash A, Savoia M,
McNama ra PJ, Schwarz CE, De Boode WP. Application of neo na-
tologist p erfor med ech ocardiography in the assessment of a pat-
ent ductus arteriosus. Pediatr Res. 2018;84:46– 56. doi: 10.103 8/
s41390- 018- 0 077- x
77. Shepherd JL, N oori S. What is a hemod ynamically signific ant PDA
in preterm i nfants? Cong enit Heart Dis. 2019;14:21– 26. doi: 1 0.1111 /
chd.12727
78. Zonnenberg I, de Waa l K. The definitio n of a haemodynamic signif icant
duct in ra ndomized contro lled trials: a system atic literature rev iew. Acta
Paediatr. 2012;101:247– 251. doi: 10 .1111/ j .16 5 1- 222 7. 2 011.0 2 46 8 . x
79. Sosenko IR, Fajardo M F, Claure N, Bancal ari E. Timing of patent duc-
tus arte riosus treatment a nd respiratory ou tcome in premature infants:
a double- blind randomized controlled trial. J Pediatr. 2012;160:929–
93 5. e9 21. doi: 1 0.1 0 16 / j .j p e d s . 2 011 .12 .0 3 1
80. Clyman RI, L iebowi tz M, Kaem pf J, Erdeve O, Bulb ul A, Hakansson S,
Lindqv ist J, Farooq i A, Kathe ria A, Sauberan J, et a l. PDA- TOLER ATE
Trial: an explor atory randomized controll ed trial of treatment of
moderate- to- la rge patent d uctus ar terios us at 1 week of age. J
Pediatr. 2019;205:41– 48. doi: 10.1016/j.jped s.2018.0 9.012
81. E rdeve O, Yurttu tan S, Altug N, Ozdemi r R, Gokme n T, Di lmen U, Oguz
SS, Uras N. Oral versu s intravenous ibuprofen for patent ductus arte-
riosus closure: a randomise d controlled trial in extrem ely low bir th-
weight infants. Arch Dis Child Feta l Neonata l Ed. 2012;97:F279 – F28 3.
doi: 10.1136/archdischild- 2011- 300532
82. Kluckow M, Evan s N. Early e chocardiographic pred iction of sy mptom-
atic patent ductus ar terios us in preter m infants undergoi ng mechani-
cal ventilation. J Ped iatr. 1995;127:774– 779.
83. Groves AM, Kusc hel CA, Knight DB, Sk inner JR. Does retrograde
diastolic flow in the descending aorta signify impaired systemic per-
fusion i n preterm infants? Pediatr Res. 2008;63:89– 94. doi: 1 0.1 2 03/
PDR.0b013e31815b4830
84. Schwarz CE, Preusche A, Baden W, Poets C F, Franz AR. Repeat abilit y
of echocardiogr aphic parameters to evaluate the h emodynamic rel -
evance of patent ductus arteriosus in preterm infants: a prospe c-
tive observational study. BMC Pediatr. 2016 ;16:18. doi: 1 0.118 6 /
s12887- 016- 05 52- 7
85. de Fre itas Martins F, Ibarra Ri os D, Resende MH, Javed H, Weisz D,
Jain A, de A ndrade Lopes JM, Mc Namara PJ. Rel ationship of patent
ductus arterio sus size to echocardiograph ic markers of shunt volume.
J Pediatr. 2018;202:50– 55.e53. doi: 10.1016/j.j pe ds .20 18.0 6.045
86. Bro adhouse KM, Price A N, Durighel G, Cox DJ, Fi nnemore AE,
Edwards AD, Hajnal JV, Groves AM. Assessment of PDA sh unt and
systemic b lood flow in newbor ns using c ardiac MR I. NMR Biomed.
2013;26:1135– 1141. doi: 10.1002 /nb m. 2927
87. Sehgal A, Paul E, M enahem S. Functional echocardiography in stag-
ing for duc tal disease sever ity: role in predicting outcomes. Eur J
Pediatr. 201 3;17 2:17 9 – 18 4 . doi: 10 .1007/s 0 04 31- 012- 1851- 0
88. Schena F, Francescato G, Cappeller i A, Picci olli I, Mayer A, Mosca
F, Fumagalli M. Association between hemodynamically significant
patent ductus arteriosus and bronchopulmonary dysplasia. J Pediatr.
201 5;16 6:14 8 8 – 149 2. doi: 10.1016/j.jpe ds.2 015.03.012
89. El- Khu ffash A, James AT, Corcoran JD, Dicker P, Franklin O, Elsayed
YN, Ting JY, Sehgal A, Malikiwi A, Harabor A , et al. A patent ductus
arteriosus severity score predicts chronic lung disease or death be-
fore discharge. J Ped iatr. 2 015;167:1354 – 13 61.e1352. doi: 10.1016/j .
jpeds. 2015.09.028
90. Andrade A, Vargas- Ba rron J, Rijlaarsd am M, Romero- Cardenas A,
Keirns C, Esp inola N. Utility of transesophagea l echocardiography in
the examination of adult patients with patent ductus arteriosus. Am
Heart J. 1995;130:543– 546. doi: 10.1016/0 002- 8703 (9 5) 90 36 4- x
91. Silver sides CK , Granton JT, Konen E, Hart MA , Webb GD, Therrien J.
Pulmonary thrombosis in adults with Eisenmenger syndrome. J Am
Coll Cardiol. 2003;42:19 82 – 1987. doi: 10.1016/j .j acc.2 00 3.0 7.022
92. Goitein O, Fuhrma n CR, Laco mis JM. Incidental finding on MDCT of
patent ductus arte riosus: use of CT and MRI to as sess clinical im-
portance. AJR Am J Roentge nol. 200 5;18 4:1924 – 19 31. d oi: 10.2 214 /
ajr.184.6.01841924
93. Zhang DZ, Zhu X Y, Lv B, Cui CS, Han XM, She ng XT, Wang QG,
Zhang P. Trial occlusio n to assess the risk of pe rsisten t pulmonary
arterial hypertension after c losure of a large patent d uctus ar te-
riosus in adolescents and adults with elevated pu lmonar y arte ry
pressure. Circ Ca rdiovasc Interv. 2014;7:473– 481. doi: 10 .116 1/
CI R C I N T E RV E N T I O NS .11 3. 0 0 113 5
94. Niu MC, Mallor y GB, Justino H, Ruiz FE, Peti t CJ. Treatment of severe
pulmonary hypertens ion in the setting of the large patent ductus arteri-
osus. Pediatrics. 2013;13 1:e16 43 e16 49 . doi: 10.1 5 42 / p e d s . 2011- 1417
95. Cooke L, Steer P, Woodgate P. Indomethaci n for asymptomatic patent
ductus arterio sus in preterm infants. Cochrane Database Syst Rev.
2003;2:CD003745.
96. Evans P, O’Reilly D, Flyer JN, S oll R, Mitra S. Indomethacin for
symptomatic patent ductus arteriosus in preterm infants. Cochrane
Database S yst Rev. 20 21;1:CD 01313 3. doi: 10 .1002 /146 51858 .
CD 01313 3.pu b2
97. Waldher r S, Fichtne r A, Beedgen B, Bru ckner T, Schaefer F, Tonshof f
B, Poschl J, Westhoff TH, We sthoff J H. Urinary acute k idney inj ury
biomarkers in very low- birth- weight infants on indomethacin for pat-
ent ductus arteriosus. Pediatr Res. 2019;85:678– 686. doi: 10.103 8/
s41390- 019- 0332- 9
98. Stavel M, Won g J, Ciesla k Z, Sherlock R, Clave au M, Shah PS. Effect
of prophylactic indomethacin administration and early feeding on
spontaneous inte stinal perfora tion in extremely l ow- birth- weight in-
fants. J Perinatol. 2017;37: 188– 19 3. doi: 10 .1 03 8 / j p . 2 01 6.1 9 6
99. Ohlsson A, S hah SS. Ibu profen for the preventi on of patent du ctus ar-
teriosu s in preterm and/or low b irth weight infa nts. Cochrane Database
Syst Rev. 2020;1:CD004213. doi: 10.10 02/14651858.CD004213.pub5
100. Ohlsson A, Shah PS. Paracetamol (acetaminophen) for patent ductus
arteriosus in preterm or low b irth wei ght infants. Cochrane Database
Syst Rev. 2018;4:CD 010061. doi: 10.1002 /146 5185 8.C D010 0 61.pu b3
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 22
Backes et al PDA: A Contemporary Perspective
101. Härkin P, Harma A, A ikio O, Valkam a M, Leskinen M, Saa rela T,
Hallman M. Paracetamol accelerates closure of the ductus a rteriosus
after premature birth: a randomized trial. J Pediatr. 2016;177:72– 77.
doi: 10.1016 /j .jpe ds.20 16.0 4.0 6 6
102. Asbagh PA, Zarkesh MR, Nili F, Sadat NS, Nae em AT. Prophy lactic
treatme nt with oral parac etamol for patent du ctus arterio sus in preterm
infants: a ra ndomize d clinical trial. Tehran Univ Med J. 2015;7 3: 86 – 92 .
103. Lie bowitz M, Kaempf J, Erdeve O, Bulbul A, Hakans son S, Lind qvist
J, Farooqi A, Katheri a A, Sauberan J, Sing h J, et al. Comparative ef-
fectiveness of drugs used to constrict the patent du ctus ar teriosu s:
a second ary an alysis of the PDA- TOLERATE trial (NCT01958320).
JPerinato l. 2019;39:599 – 6 07. d oi: 10.103 8/s 41372- 019- 0 347- 4
104. Smyth JM, C ollier PS, Darwish M, Millership JS, Halliday HL,
Petersen S, McElnay JC. Intravenou s indomet acin in preterm in-
fants with symptomatic patent ductus arteriosus. A population
pharmacokinetic study. Br J Clin Pharmaco l. 2004;58:249– 258. doi:
10 .1111/ j .13 6 5 - 21 25 . 2 0 0 4.0 2 13 9 .x
105. Nak ajima M, In oue T, Shimada N, Tokudome S, Yamamoto T, Kuroiwa
Y. Cytochrome P450 2C9 c atalyzes indom ethacin O - demethylation in
human liver microsomes. Drug Metab Dispos. 1998;26:261– 266.
106. Ma no Y, Usui T, Kamimura H. Contr ibution of UDP-
glucuronosyltransferases 1A9 and 2B7 to the glucuronidation of indo-
methacin in the huma n liver. Eur J Clin Pharmacol. 2007;63:289– 296.
doi: 10.1007/s00228 - 007- 0261- 0
107. Blake MJ, Castro L, Leeder JS, Kearns GL. Ontogeny of drug me-
tabolizing enz ymes in th e neonate. Semin Fetal Neonatal Med.
20 05;10:123 – 13 8. doi: 10 .10 16 / j .s i n y. 2 0 0 4 .11. 0 01
108. Badee J, Qiu N, Collier AC, Takahashi RH, Forre st WF, Parrott N,
Schmidt S, Fowler S. Character ization of the ontogeny of hepatic
UDP- glucuronosyltransferase enzymes based on glucuronidation
activity measured in human liver microsomes. J Clin Pharmacol.
2019;59(suppl 1):S42– S55. doi: 10 .10 02 / j cp h .14 9 3
109. Smith CJ, Ryckman KK, Bahr TM, Da gle JM. Poly morphisms in
CYP2C9 are associated with response to indomethacin among ne-
onates with patent ductus arteriosus. Pediatr Res. 2017;82:776– 780.
doi: 10.1 0 3 8/ p r. 2 0 17.14 5
110. Rooney SR, Shelton EL, Aka I, Shaf fer CM, Clyman RI, D agle JM,
Ryckma n K, Lewis TR , Reese J, Van Driest SL, et al. CYP2C9*2 is
associated with indomethacin treatment failure for patent ductus
arteriosus. Pharmacogenomics. 2019;20:939– 946. doi: 10.2 217/
pgs- 2 019- 0 079
111. Yang M- C, Liu H- K, Wu H - Y, Tey S- L, Yang Y- N, Wu C- Y, Wu J- R. Initial
experience with patent ductus arteriosus ligation in p re- term i nfants
with bidi rection al shunt pattern. Front Pediatr. 2020;8:591441. doi:
10.3389/fpe d.2020.591441
112. Peckham GJ, Miettinen OS, Ellison RC, Kray bill EN, Ge rsony WM,
Zierle r S, Nadas AS. Clinica l course to 1 year of age in premature i n-
fants with patent ductus arteriosus: results of a multicenter random-
ized trial of indomethacin. J Pediatr. 1984;105:285 – 291. do i: 10.1016/
s0022- 3476(84)80134- 1
113. G ersony WM, Peckha m GJ, Ellison RC, Miett inen OS, Nadas AS. Ef fects
of indomethacin in premature infants with patent ductus arteriosus: re-
sults of a national collaborative study. J Pediat r. 1983;102:895– 906.
114. Clyman R, Cassady G, K irklin J K, Collins M, Philips JB III. The ro le of
patent ductus arteriosus ligation in bronchopulmonary dysplasia: re-
examining a randomized controlled trial. J Pediatr. 20 09;154:873– 876.
doi: 10.1016/j. jp ed s. 2009. 01.0 05
115. Malviya MN, Ohlss on A, Shah SS. Surgic al versu s medica l treat-
ment with cyclooxygenase inhibitors for symptomatic patent duc-
tus arte riosus in preterm infants. Coc hrane Database Syst R ev.
2013;2013:C D0 039 51. doi: 10.1002/14651858.CD003951.pub3
116. Mosal li R, Alfaleh K. Prop hylactic surgica l ligatio n of patent duc tus ar-
teriosu s for prevention of mor tality and morb idity in extreme ly low bir th
weight infants. Cochra ne Database Syst Rev. 2008;2008:CD006181.
doi: 10.1002 /1465185 8. CD 00 6181. pu b2
117. American College of Obstetricians and Gynecologists, Soci ety for
Materna l- Fetal Medicine. Obstetric care consensu s no. 6: periv i-
able bir th. Obstet Gynecol. 2017;13 0:e18 7– e199 . do i: 10.1097/
AOG.0000000000002352
118. McNamara PJ, Stewar t L, Shivananda SP, Stephe ns D, Sehgal A.
Patent ductus arter iosus li gation is a ssociated with impaired lef t ven-
tricula r systolic p erfor mance in p remature infants weighing le ss than
1000 g. J Thorac Cardiovas c Surg. 2 010;14 0:15 0 – 157. doi: 10 .1016/j .
jtcvs.2010.01.011
119. Teixeira LS, Shivan anda SP, Stephens D, VanA rsdell G, McNamara
PJ. Postoperative cardiorespiratory instability following ligation of the
preterm du ctus ar teriosu s is related to e arly ne ed for inter ventio n.
JPerinato l. 2008;28:803– 810. doi: 10.1038/jp. 20 08.101
120. C horne N, Le onard C, Pie cuch R, Cl yman RI. Pate nt ductus a rteri osus
and its treatment as risk factor s for neonatal and ne urodevelopment al
morbidity. Pediatrics. 2 0 07;119: 116 5 1174.
121. Kab ra NS, Schm idt B, Robe rts RS, Doyle LW, Papile L, Fana roff A.
Neurosensory impairment after surgical closure of patent ductus ar-
teriosu s in extrem ely low birth weig ht infants: re sults fro m the Trial of
Indomethacin Prophylaxis in Preterms. J Pediatr. 2007;150:229– 234.
doi: 1 0.1 01 6 / j. j p e d s . 2 0 0 6.11 .0 3 9
122. Henry B M, Hsieh WC, Sa nna B, Vikse J, Taterra D, Tomaszewski K A.
Incidence, risk factors, and comorbidities of vocal cord paralys is after
surgical closure of a patent duc tus arte riosus: a meta- analysis. Pe diatr
Cardiol. 2019;40:116– 125. doi: 10.1007/s 00 246 - 0 18- 196 7- 8
123. Roclawski M, Pankowski R, Smoczynsk i A, Ceynowa M, Kloc W,
Wasilewski W, Jende P, Liczbik W, Beldz inski P, Libionka W, et al.
Second ary scoliosis a fter thoracotomy in patients with aorti c coarc-
tation and patent ductus arteriosus. S tud Health Technol Infor m.
20 12;17 6: 43 4 6 .
124. Backes CH, Ri vera BK, Bridge JA, Armstrong AK, Boe BA, Berman
DP, Fick T, Holzer RJ, Hijazi ZM, Abadir S, et al. Percutaneous p at-
ent ductu s arter iosus (PDA) c losure du ring infancy: a meta- anal ysis.
Pediatrics. 2017;139:e20162927. doi: 10.1542/peds.2016- 2 927
125. Barcroft M, McKee C, Berman DP, Taylor RA, R ivera BK, Smich CV,
Slaughte r JL, El-Khuffa sh A, Backes CH. Percutaneous closure of p at-
ent ductus arteriosus. Clin Perinatol. 2022;49:149– 166. doi: 10.1016/j .
c lp . 2 02 1.11 .0 0 9
126. Sathananda m SK, Gut finger D, O’Brien L, For bes TJ, Gillespie MJ,
Berman DP, Armstrong AK, Sh ahanava z S, Jones TK, Morray BH, et
al. Amplatzer Piccolo Occluder clinical trial for percutaneous closure
of the patent ductus ar teriosus in patients >/=700 grams. Cathete r
Cardiovas c Interv. 2020 ;9 6:12 66 – 12 76. doi: 10.10 02 /cc d.2 897 3
127. Backes CH, Giesinge r RE, Rivera BK, Berm an DP, Smith CV, Cua CL,
Kellehe r KJ, McNam ara PJ, Slaughte r JL. Percut aneous c losure of th e
patent duc tus arte riosus in very low weight infants: consi deratio ns fol-
lowing US Fo od and Drug Administration a pproval of a novel device.
JPediatr. 2019;213:218– 2 21. doi: 10.1016/ j.j pe ds .20 19.0 5.0 62
128. Bischoff AR, Jasani B, Sathana ndam SK, Backes C, Weisz D E,
McNama ra PJ. Percutane ous clos ure of patent d uctus ar terios us in
infants 1.5 kg or les s: a meta- anal ysis. J Ped iatr. 2021;230:84– 92.e14.
doi: 10.1016/j. jp ed s. 202 0.10.0 35
129. Fraisse A , Bautista- Rodr iguez C, Burmester M, L ane M, Sin gh Y.
Transcatheter closure of patent ductu s arteriosus in infants wi th weight
under 1,500 gra ms. Front Pediatr. 2020;8:558256. doi: 10.3389/
fped.2020.558256
130. Sathanandam S, G utfinger D, Morr ay B, Berma n D, Gillespie M,
Forbes T, Johnso n JN, Garg R, M alek zadeh- Milani S, Fra isse A,
et al. Consensus g uidelines for the p revention and management of
periprocedural complications of transcatheter patent ductus arte-
riosus closure wi th the ampl atzer piccolo occluder in extremely low
birth we ight infa nts. Pediatr Cardiol. 2021;4 2:1258 – 1274. doi: 10.1007/
s00246- 021- 02665- 3
131. de Waal K, Prasad R, Kluckow M. Patent ductu s arter iosus ma n-
agement and the drift towards therapeutic nihilism— what is th e ev-
idence? Semin Fetal Neonatal Med. 2021;26:101219. do i: 10.1016/ j.
siny.2021.101219
132. Altit G, Saeed S, Beltempo M, Claveau M, L apointe A, Basso O.
Outcome s of extrem ely prem ature infa nts compa ring patent ductus
arteriosus management approaches. J Pediatr. 2021;235:49– 57.e42.
doi: 10.1016/j. jp ed s. 2021.04. 014
133. Relangi D, Somashek ar S, Jain D, Vanbu skirk S, B ancala ri E, Sosenko
I, Claure N. C hanges in patent d uctus arter iosus treatme nt strategy and
respiratory outcomes in premature infants. J Pediatr. 2021;235:58– 62.
doi: 10.1016/j.jpeds.2021.04.030
134. Fir th J, Picker ing D. Timin g of indomethacin the rapy in persistent d uc-
tus. Lancet. 19 8 0; 2:14 4.
135. Herrman K, Bose C, Lewis K, Laughon M. Spontaneous closu re of
the patent ductus ar teriosus in very low birth weight infants following
discharge from the neonata l unit. Arch Dis Child Fetal Neonatal Ed.
2009;94:F48– F50. doi: 1 0.11 36 / a d c. 2 0 07.1 29 2 70
136. Weber SC, Weis s K, Buhre r C, Hansmann G, Koehne P, Sallmon H.
Natural history of patent ductus arter iosus in ve ry low bi rth weight
Downloaded from http://ahajournals.org by on September 3, 2022
J Am Heart Assoc. 2022;11:e025784. DOI: 10.1161/JAHA.122.025784 23
Backes et al PDA: A Contemporary Perspective
infants af ter disc harge. J Pediatr. 2 0 15 ;16 7: 114 9 – 11 51. d oi: 10.1016/ j.
jpeds. 2015.06.032
137. Tolia VN, Powers GC, Kelleher AS, Walker MW, Herrman KK, Ahm ad
KA, Buchh B, Egal ka MC, Hinkes M, Ma M, et al. Low rate of sponta-
neous closure in premature infants discharged with a patent ductus
arteriosus: a multicenter prospective study. J Pediatr. 2022;240:31–
36.e 32. doi: 10.1016/j.j pe ds .20 21.07.03 5
138. Moore J W, Greene J, Palo mares S, Javois A, Owad a CY, Cheatham
JP, Hoyer MH, Jones TK, Levi DS; Pivota l. Results of the combi ned
U.S. multice nter pivota l study an d the contin uing acc ess study of the
Nit- Occlud PDA device for percutaneous closure of patent ductus ar-
teriosus. JACC Cardiovasc Interv. 2 014;7:14 3 0 – 14 36 . doi: 10.1016/j .
jcin .2014.06. 019
139. Feltes T F, Bacha E, Be ekman RH III, Ch eatham JP, Feinstein JA, Go mes
AS, Hijazi ZM, Ing FF, de Moor M, Morrow WR, et al. In dicatio ns for
cardiac catheterization and intervention in pediatric cardiac disease: a
scienti fic statement from the American Hea rt Association. Circulation.
2011;123:2607– 2652. doi: 10 .116 1/ C I R . 0 b 01 3e 3 18 2 1b1 f 10
140. Salavitabar A, Krishnan US, Turner M E, Vincent JA, Torres AJ, Cr ystal
MA. Safet y and outc omes of tra nscatheter closure of patent duc tus
arteriosus in children w ith pulmo nary arter y hyper tension. Tex Heart
Inst J. 2020;47:250– 257. doi: 10.14503/THIJ- 19- 6982
141. Dimo poulos K, Inuzuka R, Goletto S, G iannakoulas G, Swa n L,
Wort SJ, Gatzoulis M A. Improve d surv ival among patients with
Eisenmenger syndrome receivin g advanced therapy for pulmonary
arterial hypertension. Circulation. 2010;121:20– 25. doi: 10 .116 1/
CIRCULATIONAHA.109.883876
142. Nezafati MH, Mah moodi E, Ha shemia n SH, Hamedanchi A . Video -
assisted thoracoscopic surgical (VATS) closure of patent ductus arteri-
osus: repo rt of three- hundr ed cases. Hear t Surg Forum. 200 2; 5:5 7– 5 9.
143. Mavroudis C, Backe r CL, Gevit z M. Fort y- six years of patient duc-
tus arteriosus division at Children’s Memorial Hospital of Chicago.
Standa rds for comparison. Ann Surg. 1994;220:402– 409; discussion
40 9 410. doi: 10.1097/00000658- 199409000- 00016
144. Balzer DT, Spray TL, McMullin D, Cottin gham W, Canter CE.
Endar teritis associate d with a clinically silent patent ductus arteriosus.
Am Hear t J. 1993;125:1192– 1193 . doi: 10.1016/ 00 02- 870 3( 93) 90 144- x
145. Gross RE. Complete d ivisio ns for the pate nt ductus a rteri osus.
JThorac Su rg. 19 47 ;1 6 :3 14 3 2 7.
146. Slaughter JL, Cua CL, Note stine JL, Rivera BK, M arzec L, H ade
EM, Maitre NL, Kleba noff MA , Ilgenf ritz M, Le V T, et al. Early pre -
diction of spontaneous patent ductus arteriosus (PDA) closure and
PDA- ass ociated outcomes: a prospecti ve cohor t investi gation. BMC
Pediatr. 2019;19:3 33. doi: 1 0 .118 6 /s 12 8 87- 019 - 17 0 8 - z
Downloaded from http://ahajournals.org by on September 3, 2022
... In neonates, the closure of the ductus arteriosus occurs in more than 90% of instances within 48 hours, and in all cases by 96 hours after birth. Some individuals may continue to have PDA throughout later childhood or adulthood [8]. PDA is also found with increased frequency in several genetic syndromes, including Down syndrome (trisomy 21) [8]. ...
... Some individuals may continue to have PDA throughout later childhood or adulthood [8]. PDA is also found with increased frequency in several genetic syndromes, including Down syndrome (trisomy 21) [8]. ...
... In adult patients who meet the criteria for PDA closure, percutaneous occlusion is preferred over the surgical approach, usually with ductal occluders, such as Amplatzer devices [3,4]. ...
... PDA closure is contraindicated in patients with severe and irreversible pulmonary hypertension, as in the case of patients with Eisenmenger syndrome, where PDA closure may worsen clinical symptoms and reduce the cardiac output [1,4]. ...
... Globally, PDAs occur in 1 in 2,000 births, and account for 5% to 10% of all CHD, with an incidence inversely related to gestational age and weight and directly related to high altitude. 4,5 In high-altitude dwellers such as our patient, PDAs have a tendency toward larger ductal diameters and higher PA pressures than those living at low altitude. 6 While some believe ductal patency persists due to lower arterial oxygen tension, it is still unknown whether certain phenotypic adaptations to chronic hypobaric hypoxia can lead to variable degrees of PDAs among those at high altitude. ...
... 1 ''Silent'' PDAs, diagnosed by imaging in asymptomatic patients, can be seen in approximately 1 in 20 births. 4 Our patient, who previously experienced symptoms, was not overly dyspneic or fatigued during pregnancy prior to identifying a large PDA. Pregnancy can affect the clinical expression of a PDA by modifying the ratio between the pulmonary and systemic flows primarily driven by an increase in total blood volume, since the mean PA pressure and the PVR stay relatively stable in the third trimester. ...
Article
Full-text available
Highlights •Large uncorrected PDA may first be diagnosed during pregnancy. •Multimodality imaging may be safely utilized in these patients. •Multimodality imaging is safely utilized in these patients. •Multidisciplinary team approach is key for percutaneous closure of PDA in pregnancy.
... A ventricular septal defect (VSD) is an important lesion that can present isolated or as a component of a complex congenital cardiac malformation [3] . Regarding the patent ductus arteriosus (PDA), the hemodynamic consequences are markedly variable and depend on its size and the cardiopulmonary status of the patient [4] . On the other hand, aortic arch hypoplasia can also present as an isolated abnormality or be associated with other congenital heart defects, especially coarctation of the aorta (CoA), and leads to severe cardiovascular complications if left untreated [5] . ...
Article
Full-text available
Ventricular septal defect is the most common congenital heart disease in children and is associated with patent ductus arteriosus in 1%-7% of cases. The coexistence of both malformities with hypoplastic aortic arch and aortic coarctation is even rarer. We present the case of a 6-year-old girl referred to our hospital because of dyspnea on feeding, recurrent respiratory infections, poor weight gain, and a heart murmur. The image studies revealed a ventricular septal defect, patent ductus arteriosus, severe hypoplasia of the aortic arch with critical stenosis of the proximal portion, severe dilatation of the pulmonary artery and pulmonary, mitral, tricuspid, and aortic regurgitation. We will discuss the diagnostic approach and treatment in a tertiary reference center for patients with cardiovascular diseases.
... Recent research reports indicate that >50% of infants born at gestational age <26 weeks have an open ductus after 2 months post-birth. 11 Infants born weighing less than 1000 grams are at the highest risk for PDA. In this population, 70% will develop PDA by day 7. 7 The patient's mother did not experience any complications during her pregnancy, however, one of the patient's siblings is currently undergoing pharmacological treatment for PDA diagnosis. ...
Article
Background: Patent Ductus Arteriosus (PDA) is a prevalent congenital heart defect characterized by the persistence of an open arterial connection between the aorta and the pulmonary. Percutaneous PDA closure involves deploying occlusion devices through catheter- based procedures. Case Presentation: We report a 1-year and 6-month-old girl diagnosed with Patent Ductus Arteriosus, presented with persistent complaints of failed to thrive , shortness of breath and recurrent cough. Physical examination revealed a continuous murmur at the upper left sternal border, and the Echocardiography examination confirmed a PDA with size of 4-7 mm, with diastolic shortening and dilatation of the Main Pulmonary Artery (MPA). The successful percutaneous PDA device closure procedure was performed using Three-Dimensional Rotational Angiography (3DRA), following an attempt with Two-Dimensional Angiography, which failed to determine the actual defect size in the patient. Conclusions: Confirming the morphology and size of a PDA can be difficult due to its location between the aorta and pulmonary blood vessels, making it a challenging congenital anomaly to assess accurately. 3DRA precisely delineates the PDA structure located between the aortic and pulmonary blood vessels and acts as a guidance tool for percutaneous PDA device closure.
Article
The aim of this study was to introduce an interventional heart catheterization to close patent ductus arteriosus (PDA), ventricular septal defect (VSD), atrial septal defect (ASD), and pulmonary hypertension without complications from open heart surgery and a 3-day hospitalization period. PDA, VSD, and ASD are among the most common abnormalities associated with various complications. This case is a 3.5-year-old girl with frequent lung infections and Failure to thrive. Treatment in the first stage aims to close the PDA using Amplatzer ADO II type AGA, size 5-6 mm and ASD using Amplatzer Septal Occluder size 15 mm. The patient was discharged the next day. Six months later, a successful interventional closure of the VSD was performed using Lifetech Symmetric Amplatzer membranous size 12 mm and patient was discharged 2 days after. All these defects were corrected without open heart surgery and the need for long-term Intensive care unitsadmission.
Preprint
Full-text available
Background Pulmonary vascular disease (PVD) and pulmonary hypertension (PH) is a significant disorder affecting prognosis of extremely preterm infants. However, there is still a lack of a consensus on the definition and optimal treatments of PH, and there is also a lack of research comparing these conditions with persistent pulmonary hypertension of newborn (PPHN), early PH, and late PH. To investigate pulmonary hypertension (PH) in extremely preterm infants, this study compared the basal characteristics, short-term outcomes, and treatment duration, categorized by the timing of requiring PH treatment. Methods This study retrospectively analyzed extremely preterm infants admitted to a single tertiary center. Between 2018 and 2022, infants with clinical symptoms or echocardiographic evidence of PH who required treatment were divided into three groups based on the timing of treatment initiation: from birth to day 2 (extremely early-period), from day 3 to day 27 (early-period), and after day 28 (late-period). The study compared the outcomes, including mortality rates, bronchopulmonary dysplasia (BPD) severity, PH treatment duration, and oxygen therapy duration, among the three groups. Results Among the 157 infants, 67 (42.7%) were treated for PH during their stay. Of these, 39 (57.3%) were treatment in extremely early, 21 (31.3%) in early, and seven (11.4%) in late periods. No significant differences were observed in maternal factors, neonatal factors, or morbidity between the three groups. However, infants who received extremely early-period treatment had a higher mortality rate, but shorter duration of noninvasive respiratory support, oxygen therapy, and PH medication use. On the other hand, the late-period treatment group received longer durations of respiratory support and treatment. Conclusions This study revealed differences in mortality rates, respiratory outcomes, and treatment duration between the three groups, suggesting varying pathophysiologies over time in extremely preterm infants.
Article
Full-text available
The patent ductus arteriosus frequently poses a significant morbidity in preterm infants, subjecting their immature pulmonary vascular bed to substantial volume overload. This, in turn, results in concurrent hypoperfusion to post-ductal organs, and subsequently alters cerebral blood flow. In addition, treatment has not demonstrated definitive improvements in patient outcomes. Currently, the optimal approach remains a subject of considerable debate with ongoing research controversy regarding the best approach. This article provides a comprehensive review of existing literature.
Article
Full-text available
The ductus arteriosus (DA) is a unique fetal vascular shunt, which allows blood to bypass the developing lungs in utero. After birth, changes in complex signaling pathways lead to constriction and permanent closure of the DA. The persistent patency of the DA (PDA) is a common disorder in preterm infants, yet the underlying causes of PDA are not fully defined. Although limits on the availability of human DA tissues prevent comprehensive studies on the mechanisms of DA function, mouse models have been developed that reveal critical pathways in DA regulation. Over 20 different transgenic models of PDA in mice have been described, with implications for human DA biology. Similarly, we enumerate 224 human single‐gene syndromes that are associated with PDA, including a small subset that consistently feature PDA as a prominent phenotype. Comparison and functional analyses of these genes provide insight into DA development and identify key regulatory pathways that may serve as potential therapeutic targets for the management of PDA.
Article
Full-text available
Transcatheter closure of patent ductus arteriosus (PDA) in premature infants is a feasible, safe, and an effective alternative to surgical ligation and may be performed with an implant success rate of 97%. Major procedural complications related to transcatheter PDA closure in extremely low birth weight (ELBW) infants are relatively infrequent (< 3%) ,but may be associated with a fatality if not optimally managed. Operators performing transcatheter PDA closures should be knowledgeable about these potential complications and management options. Prompt recognition and treatment are often necessary to avoid serious consequences. With strict guidelines on operator training, proctoring requirements, and technical refinements, transcatheter PDA closure in ELBW infants can be performed safely with low complication rates. This article summarizes the consensus guidelines put forward by a panel of physicians for the prevention and management of periprocedural complications of transcatheter PDA closure with the Amplatzer Piccolo Occluder in ELBW infants.
Article
Full-text available
Background: A meta-analysis published in 2015 showed a significant association between low platelet counts in the first day(s) of life and risk of patent ductus arteriosus (PDA). The meta-analysis pooled data from 11 studies cohorts (3,479 preterm infants). Objective: To update the meta-analysis by adding new studies on the topic and including other platelet parameters different from platelet counts. Methods: PubMed/Medline and Embase databases were searched. Random-effects risk ratios (RR) and differences in means (DM) and 95% confidence intervals (CI) were calculated. Results: We included 31 studies (7,638 infants). Meta-analysis showed that the risk of developing any PDA was significantly associated with platelet counts<150 × 10 ⁹ /L (11 studies, RR 1.58, 95% CI 1.28 to 1.95), and <100 x 10 ⁹ /L (7 studies, RR 1.61, 95% CI 1.14 to 2.28), but not <50 x 10 ⁹ /L (4 studies, RR 1.34, 95% CI 0.77 to 2.32). Risk of developing hemodynamically significant PDA (hsPDA) was significantly associated with platelet counts<150 x 10 ⁹ /L (12 studies, RR 1.33, 95% CI 1.09 to 1.63), and <100 x 10 ⁹ /L (7 studies, RR 1.39, 95% CI 1.06 to 1.82), but not <50 x 10 ⁹ /L (6 studies, RR 1.24, 95% CI 0.86 to 1.79). Infants with hsPDA had significantly lower mean platelet counts (19 studies, DM 22.0 x 10 ⁹ , 95% CI 14.9 to 29.1) and platelet mass (11 studies, DM 214.4, 95% CI 131.2 to 297.5) and significantly higher platelet distribution width (PDW, 9 studies, DM −0.53, 95% CI −1.01 to −0.05) than infants without hsPDA. Meta-analysis could not demonstrate significant differences in mean platelet volume (MPV). Conclusion: Compared to the previous analysis, this updated meta-analysis included 21 additional studies that provide stronger evidence of the association between low platelet counts and PDA/hsPDA. Other platelet parameters such as platelet mass and PDW are also associated with hsPDA risk. However, the low number of platelets may be an epiphenomenon associated with the maturity and clinical stability of preterm infants rather than a contributing factor in the pathogenesis of PDA.
Article
Full-text available
Background: Symptomatic patent ductus arteriosus (PDA) is associated with mortality and morbidity in preterm infants. In these infants, prophylactic use of indomethacin, a non-selective cyclooxygenase inhibitor, has demonstrated short-term clinical benefits. The effect of indomethacin in preterm infants with a symptomatic PDA remains unexplored. Objectives: To determine the effectiveness and safety of indomethacin (given by any route) compared to placebo or no treatment in reducing mortality and morbidity in preterm infants with a symptomatic PDA. Search methods: We used the standard search strategy of Cochrane Neonatal to search Cochrane Central Register of Controlled Trials (CENTRAL; 2020, Issue 7), in the Cochrane Library; Ovid MEDLINE(R) and Epub Ahead of Print, In-Process & Other Non-Indexed Citations, Daily and Versions(R); and Cumulative Index to Nursing and Allied Health Literature (CINAHL), on 31 July 2020. We also searched clinical trials databases and the reference lists of retrieved articles for randomized controlled trials (RCTs) and quasi-RCTs. Selection criteria: We included RCTs and quasi-RCTs that compared indomethacin (any dose, any route) versus placebo or no treatment in preterm infants. Data collection and analysis: We used the standard methods of Cochrane Neonatal, with separate evaluation of trial quality and data extraction by at least two review authors. We used the GRADE approach to assess the certainty of evidence for the following outcomes: failure of PDA closure within one week of administration of the first dose of indomethacin; bronchopulmonary dysplasia (BPD) at 28 days' postnatal age and at 36 weeks' postmenstrual age; proportion of infants requiring surgical ligation or transcatheter occlusion; all-cause neonatal mortality; necrotizing enterocolitis (NEC) (≥ Bell stage 2); and mucocutaneous or gastrointestinal bleeding. Main results: We included 14 RCTs (880 preterm infants). Four out of the 14 included studies were judged to have high risk of bias in one or more domains. Indomethacin administration was associated with a large reduction in failure of PDA closure within one week of administration of the first dose (risk ratio (RR) 0.30, 95% confidence interval (CI) 0.23 to 0.38; risk difference (RD) -0.52, 95% CI -0.58 to -0.45; 10 studies, 654 infants; high-certainty evidence). There may be little to no difference in the incidence of BPD (BPD defined as supplemental oxygen need at 28 days' postnatal age: RR 1.45, 95% CI 0.60 to 3.51; 1 study, 55 infants; low-certainty evidence; BPD defined as supplemental oxygen need at 36 weeks' postmenstrual age: RR 0.80, 95% CI 0.41 to 1.55; 1 study, 92 infants; low-certainty evidence) and probably little to no difference in mortality (RR 0.78, 95% CI 0.46 to 1.33; 8 studies, 314 infants; moderate-certainty evidence) with use of indomethacin for symptomatic PDA. No differences were demonstrated in the need for surgical PDA ligation (RR 0.66, 95% CI 0.33 to 1.29; 7 studies, 275 infants; moderate-certainty evidence), in NEC (RR 1.27, 95% CI 0.36 to 4.55; 2 studies, 147 infants; low-certainty evidence), or in mucocutaneous or gastrointestinal bleeding (RR 0.33, 95% CI 0.01 to 7.58; 2 studies, 119 infants; low-certainty evidence) with use of indomethacin compared to placebo or no treatment. Certainty of evidence for BPD, surgical PDA ligation, NEC, and mucocutaneous or gastrointestinal bleeding was downgraded for very serious or serious imprecision. Authors' conclusions: High-certainty evidence shows that indomethacin is effective in closing a symptomatic PDA compared to placebo or no treatment in preterm infants. Evidence is insufficient regarding effects of indomethacin on other clinically relevant outcomes and medication-related adverse effects.
Article
Percutaneous-based patent ductus arteriosus closure is technically feasible among infants less than 1.5 kg. However, marked heterogeneity in the type and nature of adverse events obscures current safety profile assessments. Although data on the risks of postdevice closure syndrome remain promising, a lack of comparative trials of surgical ductal ligation and inconsistent surveillance across published studies obscure confidence in present estimates of safety and efficacy. To minimize risk and yield the greatest benefits, clinical studies of patent ductus arteriosus treatment should consider incorporating more robust assessments to ensure that infants at greatest risk for adverse ductal consequences are included.
Article
Objectives: To assess the rate of spontaneous closure and the incidence of adverse events in infants discharged home with a patent ductus arteriosus. Study design: In a prospective multicenter study, we enrolled 201 premature infants (gestational age 23-32 weeks at birth) discharged home with a persistently patent ductus arteriosus (PDA) and followed their PDA status at 6-month intervals through 18 months of age. The primary study outcome was the rate and timing of spontaneous ductal closure. Secondary outcomes included rate of assisted closure and the incidence of serious adverse events. Results: Spontaneous ductal closure occurred in 95 infants (47%) at 12 months and 117 infants (58%) by 18 months. Seventeen infants (8.4%) received assisted closure with surgical ligation or device assisted occlusion. Three infants died (1.5%). Although infants with spontaneous closure had a higher mean birth weight and gestational age compared with infants with a persistent PDA or assisted closure, we did not identify other factors predictive of spontaneous closure. Conclusions: Spontaneous closure of the PDA occurred in slightly less than half of premature infants discharged with a patent ductus by one year, lower than prior published reports. The high rate of assisted closure and/or adverse events in this population warrants close surveillance following discharge. Trial registration ClinicalTrials.gov: NCT02750228.
Article
Objective To evaluate whether change in PDA management strategies over time had an impact on respiratory outcomes in premature infants. Study design Prospectively collected data were included from all preterm infants born at 23-30 weeks gestational age (GA) with PDA admitted to UM/JMH from January 1, 2005 to December 31, 2007 (Epoch 1) and January 1, 2011 to December 31, 2015 (Epoch 2). The 2 epochs were compared for approach to PDA diagnosis and subsequent management strategies and respiratory outcomes. Results Significantly fewer infants were treated for PDA in Epoch 2 (54%) compared with Epoch 1 (90%). Multivariable logistic regression analysis demonstrated that infants in Epoch 2, with later PDA diagnosis and less frequent PDA treatment, had greater odds of BPD, composite of BPD or death, and more treatment with postnatal steroids than in Epoch 1. Conclusion The change in approach to diagnosis and management of PDA, from a more proactive and aggressive approach during the earlier Epoch 1 to a more expectant approach during the subsequent Epoch 2, was associated with worse respiratory outcomes, including increase in BPD and in BPD or death.
Article
Objective: To evaluate the change in the proportion of deaths/bronchopulmonary dysplasia (BPD) among premature infants (<26 and 26-29 weeks) following a policy change to a strict non-intervention approach, compared with standard treatment. Study design: We examined 1249 infants (341 <26 weeks) at two comparable sites. Site #1 (control) continued medical treatment/ligation, and site #2 (exposed) changed to a non-intervention policy in late 2013. Using the difference-in-differences approach, which accounts for time-invariant differences between sites and secular trends, we assessed changes in death or BPD separately among 26-29 weeks and <26 weeks in two epochs (epoch 1: 2011-2013; epoch 2: 2014-2017). Results: Baseline characteristics were similar across sites and epochs. Medical treatment/ligation use remained stable at site #1 but declined progressively to 0% at site #2, indicating adherence to policy. We saw no difference in death/BPD among babies born at 26-29 weeks (12%, 95%CI: -1 to 24%). However, incidence of death/BPD increased by 31% among <26 weeks [95% CI: 10 to 51%] in site #2, whereas there was no change in outcomes in site #1. The Score for Neonatal Acute Physiology-Version II, used as a control outcome, did not change in either site, suggesting that our findings were not due to changes in patients' severity. Conclusion: Adherence to a strict conservative policy did not impact death or BPD among ≥26 weeks but was associated with a significant rise in infants born <26 weeks.
Article
The published literature on patent ductus arteriosus (PDA) management is challenging to interpret due to poorly designed trials with high rates of open label treatments, homogenisation of patients with varying physiological subtypes, poor treatment efficacy, and spontaneous closure in more mature infants. The perceived lack of clinical benefit has led to a drift away from medical and surgical treatment of all infants with a PDA. This therapeutic nihilism as a default response to PDA management fails to recognise the physiological relevance of a left-to-right shunt with early haemodynamic instability after birth and subsequent pulmonary volume overload with prolonged exposure. Clinicians need to know if therapeutic nihilism is safe. This review will provide an overview of the available data on the efficacy of known PDA treatments, conservative management and supportive care measures that are currently applied.
Article
To investigate whether transcatheter device closure of patent ductus arteriosus (PDA) is safe in children with pulmonary artery hypertension, we retrospectively analyzed our experience with 33 patients who underwent the procedure from January 2000 through August 2015. Pulmonary artery hypertension was defined as a pulmonary vascular resistance index (PVRI) >3 WU · m2. All 33 children (median age, 14.5 mo; median weight, 8.1 kg) underwent successful closure device implantation and were followed up for a median of 17.2 months (interquartile range [IQR], 1.0–63.4 mo). During catheterization, the median PVRI was 4.1 WU · m2 (IQR, 3.6–5.3 WU · m2), and the median mean pulmonary artery pressure was 38.0 mmHg (IQR, 25.5–46.0 mmHg). Premature birth was associated with pulmonary vasodilator therapy at time of PDA closure ( P=0.001) but not with baseline PVRI (P=0.986). Three patients (9.1%) had device-related complications (one immediate embolization and 2 malpositions). Two of these complications involved embolization coils. Baseline pulmonary vasodilator therapy before closure was significantly associated with intensive care unit admission after closure (10/12 [83.3%] with baseline therapy vs 3/21 [14.3%] without; P <0.001). Of 11 patients receiving pulmonary vasodilators before closure and having a device in place long-term, 8 (72.7%) were weaned after closure (median, 24.0 mo [IQR, 11.0–25.0 mo]). We conclude that transcatheter PDA closure can be performed safely in many children with pulmonary artery hypertension and improve symptoms, particularly in patients born prematurely. Risk factors for adverse outcomes are multifactorial, including coil use and disease severity. Multicenter studies in larger patient populations are warranted.