You are on page 1of 9

TAGEDENS E M I N A R S I N P E R I N A T O L O G Y 46 (2022) 151591

Available online at www.sciencedirect.com

Seminars in Perinatology
www.seminperinat.com

Periventricular- intraventricular hemorrhage in the


premature infant- A historical perspective
Jeffrey M. Perlman*
Department of Pediatrics, Weill Cornell Medicine, Division Chief of Newborn Medicine, New York Presbyterian Hospital, 1283 York Avenue
15th Floor, New York, NY, 10065

AB STR ACT

The objective of this chapter is to trace the evolution of intraventricular hemorrhage in the pre-
mature infant highlighting the importance of the germinal matrix, a critical role for cerebral
blood flow changes in the genesis of hemorrhage, clinical factors that increase the bleeding risk,
and potential preventative strategies. In 1976, neuropathological studies demonstrated capillary
rupture within the germinal matrix as the precursor of hemorrhage. In 1980, introduction of cra-
nial ultrasound facilitated diagnosis of intraventricular hemorrhage. In 1979, loss of cerebral
autoregulation in sick newborn infants was demonstrated. In the 1980’s, studies demonstrated
the importance of intravascular factors in provoking hemorrhage. In 1983, the association of
cerebral blood flow velocity fluctuations and subsequent hemorrhage was demonstrated. In
1994, antenatal steroids use to accelerate lung development was recommended. This was associ-
ated with an unanticipated reduction in hemorrhage. In the mid 1990’s early indomethacin
administration was associated with a reduction of severe hemorrhage.
Ó 2022 Elsevier Inc. All rights reserved.

(31%). IVH was further categorized as Grade I (blood within the


Background germinal matrix (GM)), Grade II (some blood within the lateral
ventricles), Grade III (blood filling the lateral ventricles), and Grade
Periventricular  intraventricular PV-IVH, particular when severe, IV (IVH with adjacent parenchymal hemorrhage). The smaller
remains the most common serious neurologic lesion of the neo- hemorrhages resolved spontaneously whereas hydrocephalus fre-
natal period, specifically in the tiniest population, i.e. < 28 weeks.1 quently developed in infants with the more severe hemorrhage.
A landmark study by Papile et al in the 1970’s utilized computed The introduction of cranial ultrasound imaging (CUS) towards the
tomography (CT) brain scanning to define the magnitude of the end of the 1970’s, utilizing the anterior fontanel as the window to
problem in 46 consecutive live-born infants < 1500g.2 The overall the neonatal brain, further defined the magnitude of the
incidence of IVH was 43% with hemorrhage significantly more problem.3,4 Thus, in the largest initial reported series of premature
likely to occur in those infants who died (64%) versus survivors infants < 2250g birthweight, approximately 40% exhibited some

Abbreviations: PV-IVH, Periventricular-Intraventricular Hemorrohae; CT, computerized tomography; GM, Germinal Matrix; CUS, Cranial
Untrosound Imaging; CBF, Cerebral Blood Flow; CBFV, Cerebral Blood Flow Velocity; ANS, Antenatal Steroids; GMH, Germinal Matrix
Hemorrhage; GA, Gestational Age; ACA, Anterior cerebarl artery; GFAP, Glial Acidic Fibrillary Protein; VEGF, Vascular Endothelial Growth
Factor; PVHI, Periventricular Hemorrhage Infarction; WMI, White Matter Injury; RDS, Respiratory Distress Syndrome; NIRS, Near Infrared
Spectroscopy; ABP, Arterial Blood Pressure; HMD, Hyaline Membrane Disease; CP, Cerebral Palsy; RR, Relative Risk; CI, Confidence Inter-
val; PIH, Pregnancy Indiced Hypertension
*Corresponding author.
E-mail address: JMP2007@med.cornell.edu

https://doi.org/10.1016/j.semperi.2022.151591
0146-0005/Ó 2022 Elsevier Inc. All rights reserved.
2 S E M I N A R S I N P E R I N A T O L O G Y 46 (2022) 151591

Table 1 – Historical Perspective of Events Related to Periventricular/Intraventricular Hemorrhage

 1976 - Neuropathological studies demonstrating rupture of capillaries within the germinal matrix as a precursor of intraventricular hemor-
rhage (IVH)5
 1976 - Computerized Tomography (CT) diagnosis of Intraventricular Hemorrhage (IVH)2
 1979 - 1980 - Cranial Ultrasound Imaging introduction facilitated the diagnosis of IVH3
 1979 - Initial Study demonstrating loss of Cerebral Autoregulation in sick newborn infants55
 1980 - 1983 - Studies demonstrating the importance of intravascular factors in genesis of IVH22-25
 1980  Studies involving Phenobarbital, Vitamin E, and Ethamsylate to prevent IVH37-41
 1983 - Association of Fluctuations in Cerebral blood flow Velocity (CBFV) and subsequent IVH17
 1985 - Elimination of Fluctuations in CBFV associated with a reduction in IVH32
 1994 - NIH consensus recommending Antenatal Steroids (ANS) use to accelerate lung development was subsequently associated with the
unanticipated reduction in IVH.
 1994 - Study showing the benefits of Indomethacin in the prevention of IVH44
 2000 - Studies demonstrating the association of chorioamnionitis/funisitis and IVH51
 2002 - Second Study demonstrating a reduction in severe IVH in infants receiving Indomethacin but with no effect on long term neurocog-
nitive outcome45
 2004 - Impact of full course of ANS on a reduction of IVH48
 2005 - Study demonstrating loss of autoregulation with increasing pCO2 levels (Hypercarbia)35

form of hemorrhage.3 Moreover, it became evident that most Monro5,6 (Fig. 1). The site of origin of GM hemorrhage (GMH)
cases occurred in the first 72 hours of life, and were most likely to varies with gestational age (GA) with most lesions in larger
evolve in the sick intubated premature infant with respiratory dis- infants originating over the head, whereas for infants < 28
tress syndrome (RDS). weeks it originates over the body of the caudate nucleus. The
This review will trace the evolution of understanding of IVH GM is a transitional gelatinous region that provides poor sup-
in the premature infant in terms of the importance of the ger- port for a large, immature network of blood vessels primarily
minal matrix (GM), a critical role for cerebral blood flow supplied by Heubner’s artery, a branch of the anterior cere-
changes (CBF) in the genesis of the lesion, clinical factors that bral artery (ACA).5-7 The microvasculature of the germinal
increase the risk of bleeding and potential preventative strat- matrix is fragile in part because of an abundance of angio-
egies (Table 1). genic blood vessels, which exhibit immaturity of basal lam-
ina, the latter lacking pericytes and fibronectin as well as a
lack of glial fibrillary acidic protein (GFAP) in the covering
astrocytes end feet.7 The associated elevation of vascular
The Germinal Matrix (GM) and the relationship to endothelial growth factor (VEGF) and angiopoietin-2 levels
IVH may be related to a relative hypoxia within the germinal
matrix. Venous drainage involves the terminal, choroidal,
The primary lesion in PV-IVH is bleeding from vessels within and thalamostriate veins that lead to the internal cerebral
the periventricular subependymal GM located between the vein. The blood flow then makes a U-turn in the subependy-
caudate nucleus and thalamus at the level of the foramen of mal region at the level of the foramen of Monro, the location
of origin of most hemorrhages. This unique venous anatomy
suggests that elevated venous pressure secondary to obstruc-
tion of venous drainage (as might occur with a large GMH)
may lead to venous distention with obstruction of the termi-
nal and medullary veins, and hemorrhagic rupture.5,6 Recent
data suggest that anatomic variants within subependymal
veins may predispose to IVH.8
The GM is site of active cellular proliferation and is a source
of neuronal precursors early in gestation as well as the source
of glial elements that become oligodendroglia and astrocytes
in the third trimester.7 It involutes after approximately 32
weeks and by term gestation, this region is essentially absent.
Its destruction may result in impairment of myelination,
brain growth, and subsequent cortical development.7

Fig. 1 – Pathological specimen from the brain of a premature Origin of the hemorrhage
infant who died from intraventricular hemorrhage. Note
blood in the germinal matrix (A), blood extending into the The vasculature of the GM has been difficult to categorize,
lateral ventricle which is distended (B), blood in the third because the small capillaries, venules, and arterioles are hard
ventricle (C) and fourth ventricle (D). to distinguish from one another histologically, in part
TAGEDENS E M I N A R S I N P E R I N A T O L O G Y 46 (2022) 151591 3

because of their relatively simple endothelial wall


structure.5,9 The primary vascular source of PV-IVH has not
been clearly established. Indeed, the initial descriptions sug-
gested a venous origin10 while others described it as emanat-
ing from capillary or arterial vessels.5 From studies
conducted over the years, it is most likely that PV-IVH is a
consequence of both arterial and venous perturbations.6,11
The hemorrhage, when it evolves, may be confined to the GM
region (Grade I) or it may extend and rupture into the adja-
cent ventricular system (defined as Grade II or III) depending
on the degree of blood, or it may extend into the white matter
(previously termed a Grade IV IVH or intraparenchymal echo-
genicity (IPE) but is more appropriately termed periventricu-
lar hemorrhagic infarction (PVHI))6,11,13 (Fig. 2 a). The latter
lesion, which is invariably unilateral, represents an area of
hemorrhagic necrosis of varying size within periventricular
white matter, dorsal and lateral to the external angle of the
lateral ventricle.3,6,12

Periventricular White Matter Injury (WMI)


associated with IVH

The etiology of the Grade IV IVH was unclear in the early 1980’s.
The initial hypothesis was that the intraparenchymal lesion rep-
resented an “extension” of hemorrhage from the GM or lateral
ventricle into previously normal periventricular white matter.2
However, subsequent neuropathological data indicated that the
intraparenchymal lesion represents regions of hemorrhagic
necrosis, due to impaired venous drainage.12-14 Furthermore, a
CBF study utilizing positron emission tomography indicated
extensive impairment involving the entire hemisphere in excess
of the hemorrhage visualized on CUS15 (Fig. 2b). We prefer the
term PVHI because, depending on the extent of involvement, it
more closely related to outcome. 12 The pathogenesis of the
white matter injury (WMI) associated with hemorrhage remains
unclear, but appears to be closely linked to the adjacent bleed.
Two potential pathways have been proposed. The first suggests
a direct relationship to the PV-IVH based on several clinical
observations: a) The WM lesion is always noted concurrent with
or following a large GM and /or IVH, b) The WMI is always
observed ipsilateral to the side of the larger hemorrhage when
there is bilateral involvement of the ventricular system.6,12,14
Fig. 2 – A Cranial ultrasound scan coronal view (top). Note
This consistent relationship between the GM and the WMI may
the large left-sided germinal matrix hemorrhage with some
in part be explained by the venous drainage of the deep white
blood in the lateral ventricle. Note the adjacent fan-shaped
matter (see above). A second explanation is a de novo evolution
intraparenchymal echodensity in the distribution of the
of WMI. Since both the GM and the periventricular white matter
medullary veins. B Positron Emission Tomography scan
represent border zone regions, the risk for ischemic injury is
(bottom). This scan is from the patient with the hemorrage
increased during periods of systemic hypotension, particularly in
noted above. The activity scan reflects actual cerebral blood
the face of a pressure passive cerebral circulation.16 Indeed, ele-
flow (CBF) measurements. Note that white-red- reflects
vated hypoxanthine and uric acid levels (perhaps as markers of
highest cerebral flow. Note the marked reduction in CBF on
reperfusion injury) have been observed on the first postnatal day
the left versus right hemisphere. Note the blood flow reduc-
in infants who subsequently developed white matter injury.18,19
tion involves the entire left hemisphere in excess of what
was observed on the cranial ultrasound scan.
Cerebral autoregulation and pressure-passive
circulation of systemic blood pressures. It was unknown in the late
1970’s whether autoregulation was intact or not in sick new-
Cerebral autoregulation is the intrinsic ability of the cerebral born infants. The seminal data of Lou and colleagues16
blood vessels to maintain relatively constant CBF over a range derived from 19 newborn infants with varying degrees of
4 S E M I N A R S I N P E R I N A T O L O G Y 46 (2022) 151591

Fig. 3 – Factors important in the pathogenesis of intraventricular hemorrhage. These are considered boadly as intravascular,
vascular, and extravascular factors. Intravascular factors appear to predominant (see text for details). CBF = cerebral blood
flow. IVH = intraventricular hemorrhage.

respiratory distress syndrom (RDS), studied in the first hours opportunity for linkage of simultaneous systemic and cere-
of life, were the first to indicate that CBF varied considerably bral circulation perturbations to the subsequent development
with spontaneous variations in blood pressure, suggesting of IVH.24
that autoregulation was lacking. Subsequent studies utilizing
different methods to assess CBF or cerebral blood flow veloc-
ity (CBFV) including Xenon, Near Infrared Spectroscopy
(NIRS), and Doppler have supported the concept of a pressure Pathogenesis of PV-IVH
passive cerebral circulation in the sick infant, i.e. CBF varying
directly with changes in systemic blood pressure.17,20-22This The genesis of PV-IVH is complex with predisposing factors
state places the developing brain at great risk for injury dur- including a combination of vascular, intravascular, and extravas-
ing times of hypotension and or elevated blood pressures. By cular influences1,2 (Fig. 3). Several reasons pointed to a critical
contrast, the circulation appears to be intact in the “stable” role for intravascular factors and specifically perturbations in
human preterm infant17 as well as in fetal and neonatal ani- arterial blood pressure (ABP) as a major factor in capillary rup-
mals.23 Notably, in fetal lambs, resting blood pressure has ture and hemorrhage. First, the cerebral circulation of the sick
been shown to be slightly above the lower limit of the autore- infant was shown to be pressure passive (see above).16,17,20-22
gulatory curve.23 This is highly relevant because if a similar Second, in a beagle puppy model, GMH could be produced by
situation exists in newborn infants, moderate hypotension induced systemic hypertension with or without prior
will result in reduced CBF and the potential for ischemic cere- hypotension.25,26 Third, infants with lower mean ABP in the first
bral ischemic brain injury. postnatal days as well infants who received rapid volume expan-
sion to correct hypotension were more likely to develop IVH.27,28
Conversely, elevations in venous pressure were also considered
Evaluation of the cerebral circulation using to be an important mechanism of hemorrhage, in part based on
Doppler the peculiarity of the anatomy of the venous drainage of white
matter and germinal matrix (see above).6,8,29 In this regard,
Application of the Doppler technique in the early 1980’s to simultaneous increases in venous pressure were observed in
measure CBFV through the anterior fontanel facilitated the infants who exhibited variability in ABP, such as with RDS and
ability to link systemic events to the pathogenesis of PV- associated complications, e.g. pneumothorax.20,29,30 Subsequent
IVH.24 Measurements were obtained from the pericallosal evidence suggested that these intravascular responses may be
artery (terminal branch of the ACA) as it courses around the modulated by additional factors, i.e. inflammation associated
genu of the corpus callosum, as well as from the right or left with chorioamnionitis.31
middle cerebral artery through the coronal sutures. Although Additional contributing factors include vascular and extra-
Doppler does not measure CBF, it has certain advantages; vascular influences, i.e. the poorly supported blood vessels,
specifically, it is noninvasive and readily available at the bed- fragility of the germinal matrix vasculature, excessive fibrino-
side and facilitates repeated measurements. When coupled lytic activity noted within the matrix region, and a prominent
with frequent sonographic assessments, it provides an postnatal decrease in tissue pressure.7,32
TAGEDENS E M I N A R S I N P E R I N A T O L O G Y 46 (2022) 151591 5

Linking Systemic and Cerebral Vascular


Perturbations in Infants with Respiratory Distress
Syndrome (RDS) to PV-IVH

Perturbations on the Arterial Side

Numerous studies had established an association between


RDS and IVH by the early 1980’s.11,33 However, the mecha-
nism(s) that linked these two co-morbidities were unclear.
Utilizing Doppler, we described an association between CBFV
fluctuations and subsequent PV-IVH in preterm infants with
RDS.20 In a study of preterm infants weighing < 1500 grams
requiring mechanical ventilation for RDS, 21 of 23 infants
(91%), with continuous beat to beat fluctuations in CBFV,
measured from around 12 hours of life, subsequently devel-
oped IVH, typically within the following 24-hours, versus 7 of
27 infants (26%) with stable CBFV patterns (Fig. 4 a, b). The
fluctuations in CBFV directly reflected similar fluctuations in
ABP in a pressure passive manner. We subsequently demon-
strated that the fluctuations were related to the respiratory
effort of the infant.34 Specifically, as work of breathing
increases, concomitant with progressive RDS, wide beat-to-
beat swings in arterial as well as venous pressures are
observed. To test the hypothesis that there was a relationship
between the systemic and cerebral fluctuations, the infant’s
respiratory effort, and development of PV-IVH, we conducted
a randomized study and eliminated the CBFV fluctuations
with muscle paralysis (Fig. 4c). All 10-control infants with
CBFV fluctuations subsequently developed IVH versus only 5
of 14 infants treated with muscle paralysis. In 4 of these 5
cases, the hemorrhage developed after cessation of the paral-
ysis.35 Importantly over time, the advent of newer ventilators
that work in tandem with the infants own respiratory efforts
coupled with the antenatal use of steroids (ANS), has made Fig. 4 – a. Stable pattern of Cerebral Blood Flow Velocity (top
this association less prominent. However, in susceptible panel) and Arterial Blood Pressure (bottom panel.). Note
infants (partial or no ANS) the same risk as described above minimal beat to beat cerebral and systemic wave form vari-
may still be apparent (see below). ability b. Beat to beat fluctuations in CBFV (top panel) and
simultaneous beat to beat fluctuations in Arterial Blood
Pressure wave form (bottom panel)in a pressure passive
Perturbations in Venous Pressure
manner. c. Note the elimination of the beat to beat fluctua-
tions in the CBFV pattern (top panel) following muscle
In subsequent studies, we observed fluctuations in venous
paralysis (bottom panel)
pressure simultaneously with ABP perturbations in preterm
infants with RDS who developed PV-IVH.29 The fluctuations
was that it had been shown that surfactant administration
can be exacerbated by higher mean airway pressures such as
acutely affects cerebral and systemic hemodynamics.37
may occur with high-frequency oscillatory ventilation, or
pneumothorax. The potential importance of these venous
fluctuations in the genesis of hemorrhage is intertwined with Hypercarbia and intraventricular hemorrhage
the venous drainage of the deep white matter (see above).
Based on experimental studies, an important role for hyper-
carbia in the genesis of IVH had been presumed. The associ-
Surfactant administration and PV-IVH ated was delineated in several neonatal studies. Thus in a
study undertaken in the first week of life in VLBW infants,
The introduction of surfactant administration to reduce the increasing PaCO2 values resulted in an increase in CBF and
severity of RDS in the late 1980’s was the most important devel- progressive impairment of cerebral autoregulation.38 Hyper-
opment in the management of sick premature infants. Surfac- capnia defined by a maximum PaCO2 recorded during the first
tant reduced mortality, severity of RDS as well as the rates of 3 days of life was associated with severe IVH. Moreover, in a
pneumothorax.36 However, in the many subsequent surfactant retrospective cohort study of 574 VLBW infants born between
trials irrespective of design, i.e. prophylactic versus rescue, syn- 1999 and mid-2004, as the maximum PaCO2 increased from
thetic versus natural, IVH persisted.36 One potential explanation 40 to 100 mm Hg, the probability of severe IVH increased from
6 S E M I N A R S I N P E R I N A T O L O G Y 46 (2022) 151591

8% to 21%.39 These cumulative observations indicate that from 14.5% to 9.0%.50 Both reports did not account for the
extremes in PaCO2 should be avoided during the period in number of doses of ANS administered to the mother.49,50 This
which infants are at high risk of IVH. is important since we demonstrated a dose dependent
decrease in IVH following a full course of ANS versus partial
or no ANS51 (Table 2). The mechanism(s) whereby glucocorti-
Strategies to prevent severe IVH  historical coids reduce severe IVH remain unclear, but may relate to
perspective (Table 1) less severe RDS, higher resting ABP or via accelerated matura-
tion of the germinal matrix region.52,53
In the early 1980’s postnatal medications used to prevent
hemorrhage included phenobarbital,40,41, Vitamin E,42,43 and
ethamyslate.44 While there was initial enthusiasm for the use Magnesium sulfate
of these medications, this was not borne out over time.
Importantly, in one large, randomized study, infants who Initial studies suggested that magnesium sulfate used to treat
received phenobarbital exhibited a higher incidence of any women with pregnancy induced hypertension (PIH) was asso-
IVH when compared to controls, i.e. 51/145 versus 26/135 ciated with a reduction in IVH.54 Subsequent studies failed to
(p=0.003) respectively.41 Another intervention that seemed show a reduction in IVH.55,56 Although a neuroprotective
promising was indomethacin. This was based on plausible effect on IVH is unclear, when magnesium sulfate is given to
experimental studies in the beagle puppy.45,46 When applied mothers at risk for preterm birth, it has been shown to reduce
clinically, the early postnatal administration of indomethacin the risk of cerebral palsy (CP) in the child in two meta-analy-
was associated with a significant reduction in severe IVH, i.e. sis. In the first report there was a reduction in CP, i.e. relative
1/209 versus 10/222 in the placebo (p=0.03).47 In a subsequent risk (RR): 0.68, (95% CI: 0.54-0.87); this comprised 5 trials total-
randomized study, Schmidt et al48 demonstrated a significant ing 6145 infants. In the second analysis, there was an almost
reduction in severe IVH (9 vs 13%) (p=0.02) in the indometha- identical result, i.e. RR 0.68, 95% CI 0.54 to 0.87, 4,601 babies, 5
cin versus placebo groups respectively. However, at 18-month trials, NNT to benefit 46.57,58
follow-up there were no differences in the incidence of cere-
bral palsy or severe cognitive delay in the two groups.48 One
hypothesis for this unanticipated observation is while indo- Pregnancy Induced Hypertension (uced
methacin reduced IVH, it had no impact on the larger under- Hypertension (PIH)
lying ischemic lesion12,15 (see above) (Fig. 2b).
PIH has been associated with a lower incidence of IVH. Thus
we reported less severe PV-IVH in infants born to mothers with
Antenatal steroids and IVH PIH than in those without PIH, i.e., 8.2% vs. 14% with an odds
ratio estimate of 0.43 (95% CI 0.30-0.61).59 Since all mothers in
The single most important “intervention” shown to signifi- this report received magnesium sulfate, it was unclear whether
cantly reduce the development of IVH has been a short course the reduction in IVH was related to the PIH or magnesium.
of antenatal glucocorticoid steroid (ANS) administered to aug- However, in another study involving mothers with PIH who did
ment pulmonary maturation.49,50 In a recent Cochrane not receive magnesium, severe IVH was reduced almost 60% in
review, a reduction in IVH was observed (average Risk Ratio newborns born to mothers with PIH versus those without PIH.60
was 0.55, 95% Confidence Interval (CI) 0.40-0.76) following any
ANS exposure.49 Importantly, this was a serendipitous obser-
vation, i.e. there has not been a prospective randomized Route of delivery
study that has evaluated the impact of ANS on reducing
severe IVH. In a more recent population-based study There is conflicting data regarding the route of delivery and
(n=28252), as ANS exposure increased from 80% in 2005 to subsequent IVH. Interpretation of the data is difficult since
90% in 2016, there was a concomitant decrease in severe IVH most studies are retrospective and undertaken before the use

Table 2 – Association of ANS Dosing in Very Low Birthweight Infants and Clinical Outcome

Antenatal Steroid Doses

Outcome None n (%) Partial n (%) Complete n (%)


HMD 37 (79) 18 (58) 18 (39) y
Surfactant 34 (72) 20 (64) 16 (35) yz
Total IVH 27 (64) 16 (52) 11 (25) yz
IVH (3 or 4) 18 (43) 8 (26) 5 (11) y
Death 18 (38) 5 (16) 6 (13) y

y p<0.05 for complete versus none.


z p<0.05 for complete versus partial.
Adapted from Reference 48
TAGEDENS E M I N A R S I N P E R I N A T O L O G Y 46 (2022) 151591 7

Table 3 – Identification of Infants Who Are At Highest


Risk for the Development of Severe Intraventricular Preventing IVH in 2021
Hemorrhage
From the above it should be apparent that a great deal has
Perinatal Factors -Minimal intrapartum care
been learned regarding the pathogenesis and as a result,
-No glucocorticoid exposure
strategies to prevent or reduce IVH, and in particular severe
-Chorioamnionitis/funisitis
IVH. Infants with severe IVH are at greatest risk for marked
Postnatal Factors -Decreasing Gestational Age neurodevelopmental deficits.12 The risk factors for severe IVH
-Decreasing Birth Weight include perinatal factors such as late arrival to the delivery
-RDS- particularly in the absence of a full room particularly during off peak hours,63 no or minimal ANS
course of glucocorticoid exposure exposure and evidence of chorioamnionitis/funisitis. Postna-
-Respiratory morbidity, i.e. pneumothorax
tal factors include very low birth weight infants < 1000 grams
-Fluctuations or rapid elevations in systemic BP
and/or GA < 28 weeks, intubated infants, and in the absence
and/or cerebral blood flow particularly in the
absence of a full course of glucocorticoid of ANS exposure, RDS, and complications, i.e. pneumotho-
exposure rax.64 (Tables 3)
-Sudden and repeated increases in venous For such infants, the risk for IVH is even greater when there
pressure are associated perturbations in arterial and venous
pressures20,29 (Fig. 3). By contrast, the risk for severe IVH in
the non-intubated infant is low, i.e., <10%.64
Our specific approach to the prevention of IVH takes into
of antenatal steroids.11 However, this does not exclude the pos-
account the exposure to ANS. Thus for infants < 1000g or <
sibility that under certain circumstances, intrapartum events
28 weeks with no or minimal ANS exposure, we administer
may contribute to the pathogenesis of severe IVH. Thus, evi-
indomethacin within six hours. The initial results of this
dence of inflammation and in particular fetal vasculitis,
approach is shown in Table 4, which shows a significant
increases the risk of IVH which may supersede the influence of
reduction in IVH with this approach.
the route of delivery.61 In one study, while vaginal delivery was
associated with an increased risk of IVH by univariate analysis,
the risks attributable to vaginal delivery were no longer ele-
vated when adjustments were made in multivariate analysis Conclusions
for fetal vasculitis and other potential confounders.61
The overall incidence of IVH including severe IVH has been
reduced over time. The most important factor contributing to
this finding has been the perinatal administration of a com-
plete course of ANS. Future strategies should target those
infants at highest risk for IVH including the severe lesion
Timing of cord clamping (Tables 3, 4). One potential strategy is the early postnatal use
of Indomethacin as described above. Additional strategies
Many reports have evaluated the potential benefit of early should be explored. Importantly, although the incidence of
versus delayed cord clamping and the impact on the develop- severe IVH has been reduced, the long-term neurodevelop-
ment of severe IVH. In the most recent and extensive review mental outcome ofinfants with lesser hemorrhage remains
on the topic involving 14 trials and totally 2972 subjects, it unclear but important to delineate.
was not possible to exclude benefit or harm from delayed
compared to early cord clamping on the occurrence of severe
IVH (RR 0.98, 95% CI 0.67 to 1.42).62 Disclosure

Table 4 – Impact of Prophylactic Indomethacin on Devel- The author report no proprietary or commercial interest in
opment of Severe IVH in Infants not Exposed to ANS at any product mentioned or concept discussed in this article.
Cornell, NYC There are no competing interests to disclose. There are no
personal or financial relationships to disclose.
Intervention # of Infants Severe IVH

Optimal ANS 55 0
R E F E R E N CE S
Suboptimal/No ANS, 9 6*
No INDO
Suboptimal/No 13 2
ANS, + INDO 1. Stoll BJ, Hansen NI, Bell EF, et al. Eunice Kennedy Shriver
National Institute of Child Health and Human Development
Neonatal Research Network. Trends in Care Practices, Mor-
Comparison of Suboptimal Antenatal Steroids (ANS)/No Indometh-
bidity, and Mortality of Extremely Preterm Neonates, 1993-
acin versus Suboptimal ANS, + Indomethacin (p=0.03)
2012. JAMA. 2015;8(10):1039–1051. https://doi.org/10.1001/
Infants who received Indomethacin vs no treatment were of # GA
jama.2015.10244: 314PMID26348753PMCIDPMC4787615.
23.8 §0.83 vs 27 §1.8 weeks (p<0.05) and BW 652 §104 grams vs
2. Papile LA, Burstein J, Burstein R, Koffler H. Incidence and evo-
972 §259 grams (p<0.05).
lution of subependymal and intraventricular hemorrhage: a
study of infants with birth weights less than 1,500 gm. J
8 S E M I N A R S I N P E R I N A T O L O G Y 46 (2022) 151591

Pediatr. 1978;92(4):529–534. https://doi.org/10.1016/s0022-3476 22. Soul JS, Hammer PE, Tsuji M, et al. Fluctuating pressure-pas-
(78)80282-0: PMID305471. sivity is common in the cerebral circulation of sick premature
3. Volpe JJ. Anterior fontanel: window to the neonatal brain. J infants. Pediatric Research. 2007;61(4):467–473.
Pediatr. 1982;00(3):395–398. https://doi.org/10.1016/s0022-3476 23. Papile LA, Rudolph AM, Heymann MA. Autoregulation of cere-
(82)80437-x: PMID7062170. bral blood flow in the preterm fetal lamb. Pediatr Res.
4. McMenamin JB, Shackelford GD, Volpe JJ. Outcome of neona- 1985;19(2):159–161.
tal intraventricular hemorrhage with periventricular echo- 24. Perlman JM. Neonatal cerebral blood flow velocity measure-
dense lesions. Ann Neurol. 1984;15(3):285–290. https://doi.org/ ment. Clin Perinatal. 1985;12:179–193.
10.1002/ana.410150315: PMID6721450. 25. Goddard-Finegold J, Armstrong D, Zeller RS. Intraventricular
5. Hambleton G, Wigglesworth JS. Origin of intraventricular hae- hemorrhage, following volume expansion after hypovolemic
morrhage in the preterm infant. Arch Dis Child. 1976;51(9):651– hypotension in the newborn beagle. J Pediatr. 1982;100(5):796–
659. 799.
6. Volpe JJ. Neurology of the Newborn. Sixth Edition. 2018. 26. Ment LR, Stewart WB, Duncan CC, Lambrecht R. Beagle puppy
7. Ballabh P. Intraventricular hemorrhage in premature infants: model of intraventricular hemorrhage. J Neurosurg.
mechanism of disease. Pediatr Res. 2010 Jan;67(1):1–8. https:// 1982;57(2):219–223.
doi.org/10.1203/PDR.0b013e3181c1b176: PMID19816235PM- 27. Bada HS, Korones SB, Perry EH, et al. Mean arterial blood pres-
CIDPMC2799187. sure changes in premature infants and those at risk for intra-
8. Tortora D, Severino M, Malova M, Parodi A. Morana Get al. Dif- ventricular hemorrhage. J Pediatr. 1990;117(4):607–614.
ferences in subependymal vein anatomy may predispose pre- 28. Goldberg RN, Chung D, Goldman SL, Bancalari E. The associa-
term infants to GMH-IVH. Arch Dis Child Fetal Neonatal Ed. 2018 tion of rapid volume expansion and intraventricular hemor-
Jan;103(1):F59–F65. https://doi.org/10.1136/archdischild-2017- rhage in the preterm infant. J Pediatr. 1980;96(6):1060–1063.
312710: Epub 2017 Jun 6PMID28588126. 29. Perlman JM, Volpe JJ. Are venous circulatory abnormalities
9. Pape KE, Wiggelsworth JS. Haemorrhage, Ischemia and the Peri- important in the pathogenesis of hemorrhagic and/or ische-
natal brain. Philadelphia, PA: JB Lippincott; 1979. mic cerebral injury? Pediatrics. 1987;80(5):705–711.
10. Larroche JC. H‘emorragies C‘er‘ebrales Intra-Ventriculaires 30. Hill A, Perlman JM, Volpe JJ. Relationship of pneumothorax to
Chez Pr‘ematur‘e. I. Anatomie et Physiopathologie. (Intraven- occurrence of intraventricular hemorrhage in the premature
tricular Cerebral Hemorrhages in the Premature. I. Anatomy newborn. Pediatrics. 1982;69(2):144–149.
and Physiopathology). Biol Neonat. 1964;7:26–56: French- 31. Yanowitz TD, Potter DM, Bowen A, Baker RW, Roberts JM. Var-
PMID14268948. iability in cerebral oxygen delivery is reduced in premature
11. Yap V, Perlman JM. Intraventricular Hemorrhage and White neonates exposed to chorioamnionitis. Pediatric Research.
Matter Injury in the Preterm Infant. In: Perlman JM, Cilio MR, 2006;59(2):299–304.
et al., eds. Neurology: Neonatology Questions and Controversies, 32. Takashima S, Tanaka K. Microangiography and fibriolytic
3rd edition. Saunders; 2019. activity in the matrix of the premature brain. Brain Dev.
12. Guzzetta F, Shackelford GD, Volpe S, Perlman JM, Volpe JJ. 1972;4:222–228.
Periventricular intraparenchymal echodensities in the pre- 33. Shalak L, Perlman JM. Hypoxic-Ischemic Cerebral Injury-Cur-
mature newborn: critical determinant of neurologic outcome. rent Concepts. Early Human Development. 2004;80:125–141.
Pediatrics. 1986;78(6):995–1006: PMID3537951. 34. Perlman JM, Thach BT. Respiratory origin of fluctuations in
13. Gould SJ, Howard S, Hope PL, Reynolds EO. Periventricular arterial blood pressure in premature infants with respiratory
intraparenchymal cerebral haemorrhage in preterm distress syndrome. Pediatrics. 1988;81:399–403.
infants: the role of venous infarction. J Pathol. 35. Perlman JM, Goodman S, Kreusser KL, Volpe JJ. Reduction in
1987;151(3):197–202. intraventricular hemorrhage by elimination of fluctuating
14. Shalak L, Perlman JM. Hemorrhagic-ischemic cerebral injury cerebral blood-flow velocity in preterm infants with respira-
in the preterm infant: current concepts. Clin Perinatol. tory distress syndrome. N Engl J Med. 1985;312:1353–1357.
2002;29(4):745–763. 36. Jobe AH. Pulmonary surfactant therapy. N Engl J Med.
15. Volpe JJ, Herscovitch P, Perlman JM, Raichle ME. Positron 1993;328(12):861–868. https://doi.org/10.1056/
emission tomography in the newborn: extensive impairment NEJM199303253281208: PMID8441430.
of regional cerebral blood flow with intraventricular hemor- 37. Kaiser JR, Gauss CH, Williams DK. Surfactant administration
rhage and hemorrhagic intracerebral involvement. Pediatrics. acutely affects cerebral and systemic hemodynamics and gas
1983;72(5):589–601: PMID6605514. exchange in very-low-birth-weight infants. J Pediatr.
16. Lou HC, Lassen NA. Friis-Hansen B. Impaired autoregulation 2004;144(6):809–814. https://doi.org/10.1016/j.
of cerebral blood flow in the distressed newborn infant. J. jpeds.2004.03.022: PMID15192631.
Pediatr. 1979;94:118–121. 38. Kaiser JR, Gauss CH, Pont MM, Williams DK. Hypercapnia dur-
17. Pryds O, Greisen G, Lou H. Friis-Hansen B. Heterogeneity of ing the first 3 days of life is associated with severe intraven-
cerebral vasoreactivity in preterm infants supported by tricular hemorrhage in very low birth weight infants. J
mechanical ventilation. J Pediatr. 1989;115(4):638–645. Perinatol. 2006;26(5):279–285.
18. Perlman JM, Risser R. Relationship of uric acid concentrations 39. Fabres J, Carlo WA, Phillips V, Howard G, Ambalavanan N.
and severe intraventricular hemorrhage/leukomalacia in the Both extremes of arterial carbon dioxide pressure and the
premature infant. J Pediatr. 1998;132:436–439. magnitude of fluctuations in arterial carbon dioxide pressure
19. Russell GAB, Jeffers G, Cook RWI. Plasma hypoxanthine: a are associated with severe intraventricular hemorrhage in
marker for hypoxic-ischemic induced periventricular leuko- preterm infants. Pediatrics. 2007;119:299–305.
malacia? Arch Dis Child. 1992;67:388–392. 40. Donn SM, Roloff DW, Goldstein GW. Prevention of intraven-
20. Perlman JM, McMenamin JB, Volpe JJ. Fluctuating cerebral tricular haemorrhage in preterm infants by phenobarbitone.
blood-flow velocity in respiratory-distress syndrome. Relation A controlled trial. Lancet. 1981;2(8240):215–217. https://doi.org/
to the development of intraventricular hemorrhage. N Engl J 10.1016/s0140-6736(81)90470-0: PMID6114279.
Med. 1983;309:204–220. 41. Kuban KC, Leviton A, Krishnamoorthy KS, Brown ER, Teele RL,
21. Tsuji M, Saul JP, du Plessis A, et al. Cerebral intravascular oxy- Baglivo JA, Sullivan KF, Huff KR, White S, Cleveland RH, et al.
genation correlates with mean arterial pressure in critically ill Neonatal intracranial hemorrhage and phenobarbital. Pediat-
premature infants. Pediatrics. 2000;106(4):625–632. rics. 1986;77(4):443–450: PMID3515304.
TAGEDENS E M I N A R S I N P E R I N A T O L O G Y 46 (2022) 151591 9

42. Chiswick ML, Johnson M, Woodhall C, et al. Protective effect 55. Leviton A, Paneth N, Susser M, Reuss ML, Allred EN, Kuban K,
of vitamin E (DL-alpha-tocopherol) against intraventricular Sanocka U, Hegyi T, Hiatt M, Shahrivar F, Van Marter LJ.
haemorrhage in premature babies. Br Med J (Clin Res Ed). Maternal receipt of magnesium sulfate does not seem to
1983;287(6385):81–84. https://doi.org/10.1136/bmj.287.6385.81: reduce the risk of neonatal white matter damage. Pediatrics.
PMID6407714PMCIDPMC1548339. 1997 Apr;99(4):E2. https://doi.org/10.1542/peds.99.4.e2:
43. Sinha S, Davies J, Toner N, Bogle S, Chiswick M. Vitamin E sup- PMID9099777.
plementation reduces frequency of periventricular haemor- 56. Perlman JM. Antenatal glucocorticoid, magnesium exposure,
rhage in very preterm babies. Lancet. 1987;1(8531):466–471. and the prevention of brain injury of prematurity. Semin
44. Morgan ME, Benson JW, Cooke RW. Ethamsylate reduces the Pediatr Neurol. 1998;5(3):202–210. https://doi.org/10.1016/
incidence of periventricular haemorrhage in very low birth- s1071-9091(98)80035-1: PMID9777678.
weight babies. Lancet. 1981;2(8251):830–831. https://doi.org/ 57. Doyle LW, Crowther CA, Middleton P, Marret S, Rouse D. Mag-
10.1016/s0140-6736(81)91103-x: PMID6116952. nesium sulphate for women at risk of preterm birth for neuro-
45. Ment LR, Stewart WB, Duncan CC, et al. Beagle puppy model protection of the fetus. Cochrane Database Syst Rev. 2009(1):
of intraventricular hemorrhage. J Neurosurg. 1982;57:219–223. CD004661. https://doi.org/10.1002/14651858.CD004661.pub3
46. Ment LR, Stewart WB, Ardito TA, Huang E, Madri JA. Indo- PMID19160238.
methacin promotes germinal matrix microvessel maturation 58. Crowther CA, Middleton PF, Voysey M, Askie L, Duley L, Pryde
in the newborn beagle pup. Stroke. 1992;23(8):1132–1137. PG, Marret S, Doyle LW, AMICABLE Group. Assessing the neu-
https://doi.org/10.1161/01.str.23.8.1132: PMID1636188. roprotective benefits for babies of antenatal magnesium sul-
47. Ment LR, Oh W, Ehrenkranz RA, et al. Low-dose indomethacin phate: An individual participant data meta-analysis. PLoS
and prevention of intraventricular hemorrhage: a multicenter Med. 2017;14(10):e1002398. https://doi.org/10.1371/journal.
randomized trial. Pediatrics. 1994;93(4):543–550: PMID8134206. pmed.1002398: PMID28976987PMCIDPMC5627896.
48. Schmidt B, Davis P, Moddemann D, et al. Long-term effects of 59. Perlman JM, Riser RC, Gee JB. Pregnancy-induced hyperten-
indomethacin prophylaxis in extremely-low-birth-weight sion and reduced intraventricular hemorrhage in preterm
infants. N Engl J Med. 2001;344(26):1966–1972. infants. Pediatr Neurol. 1997;17:29–33.
49. Roberts D, Brown J, Medley N, Dalziel SR. Antenatal cortico- 60. Murphy DJ, Sellers S, MacKenzie IZ, Yudkin PL, Johnson AM.
steroids for accelerating fetal lung maturation for women at Case-control study of antenatal and intrapartum risk factors
risk of preterm birth. Cochrane Database Syst Rev. 2017;3(3): for cerebral palsy in very preterm singleton babies. Lancet.
CD004454. https://doi.org/10.1002/14651858.CD004454.pub3. 1995;346(8988):1449–1454. https://doi.org/10.1016/s0140-6736
50. Gould JB, Bennett MV, Phibbs CS, Lee HC. Population Improve- (95)92471-x: PMID7490990.
ment Bias Observed in Estimates of the Impact of Antenatal 61. Hansen A, Leviton A. Labor and delivery characteristics and
Steroids to Outcomes in Preterm Birth. J Pediatr. 2021;232:17– risks of cranial ultrasonographic abnormalities among very-
22. https://doi.org/10.1016/j.jpeds.2020.11.067: e2Epub 2020 low-birth-weight infants. The Developmental Epidemiology Net-
Dec 1PMID33275981. work Investigators. Am J Obstet Gynecol. 1999;181(4):997–1006.
51. Salhab WA, Hynan LS, Perlman JM. Partial or complete ante- https://doi.org/10.1016/s0002-9378(99)70339-x: PMID10521768.
natal steroids treatment and neonatal outcome in extremely 62. Seidler AL, Gyte GML, Rabe H, Dıaz-Rossello JL, Duley L,
low birth weight infants < or =1000 g: is there a dose-depen- INTERNATIONAL LIAISON COMMITTEE ON RESUSCITATION
dent effect? J Perinatol. 2003;23(8):668–672. https://doi.org/ NEONATAL LIFE SUPPORT TASK FORCE. Umbilical Cord Man-
10.1038/sj.jp.7211007: PMID14647166. agement for Newborns <34 Weeks' Gestation: A Meta-analy-
52. Ballabh P, Hu F, Kumarasiri M, Braun A, Nedergaard M. Devel- sis. Pediatrics. 2021;147(3):e20200576. https://doi.org/10.1542/
opment of tight junction molecules in blood vessels of germi- peds.2020-0576: PMID33632931PMCIDPMC7924139.
nal matrix, cerebral cortex, and white matter. Pediatr Res. 63. Jensen EA, Lorch SA. Association between Off-Peak Hour Birth
2005;58:791–798. and Neonatal Morbidity and Mortality among Very Low Birth
53. Perlman JM. Glial acidic protein mRNA expression is human Weight Infants. J Pediatr. 2017 Jul;186:41–48. https://doi.org/
astroglial cells modulated by by dexamethasone. Ann Neurol. 10.1016/j.jpeds.2017.02.007: e4Epub 2017 Mar
1995;38:495. 8PMID28284476PMCIDPMC5500004.
54. Kuban KC, Leviton A, Pagano M, Fenton T, Strassfeld R, 64. Perlman JM. White matter injury in the preterm infant: an
Wolff M. Maternal toxemia is associated with reduced inci- important determination of abnormal neurodevelopment
dence of germinal matrix hemorrhage in premature outcome. Early Hum Dev. 1998;53(2):99–120. https://doi.org/
babies. J Child Neurol. 1992 Jan;7(1):70–76. https://doi.org/ 10.1016/s0378-3782(98)00037-1: PMID10195704.
10.1177/088307389200700113: PMID1552156.

You might also like