You are on page 1of 21

Handbook of Clinical Neurology, Vol.

162 (3rd series)


Neonatal Neurology
L.S. de Vries and H.C. Glass, Editors
https://doi.org/10.1016/B978-0-444-64029-1.00010-2
Copyright © 2019 Elsevier B.V. All rights reserved

Chapter 10

Neonatal encephalopathy and hypoxic–ischemic encephalopathy


ALISTAIR J. GUNN1* AND MARIANNE THORESEN2,3
1
Departments of Physiology and Paediatrics, University of Auckland, Auckland, New Zealand
2
Department of Physiology University of Oslo, Oslo, Norway
3
Neonatal Neuroscience, Translational Health Sciences, University of Bristol, Bristol, United Kingdom

Abstract
Acute hypoxic–ischemic encephalopathy around the time of birth remains a major cause of death and life-
long disability. The key insight that led to the modern revival of studies of neuroprotection was that, after
profound asphyxia, many brain cells show initial recovery from the insult during a short “latent” phase,
typically lasting approximately 6 h, only to die hours to days later after a “secondary” deterioration char-
acterized by seizures, cytotoxic edema, and progressive failure of cerebral oxidative metabolism. Studies
designed around this framework showed that mild hypothermia initiated as early as possible before the
onset of secondary deterioration and continued for a sufficient duration to allow the secondary deteriora-
tion to resolve is associated with potent, long-lasting neuroprotection. There is now compelling evidence
from randomized controlled trials that mild to moderate induced hypothermia significantly improves sur-
vival and neurodevelopmental outcomes in infancy and mid-childhood.

INTRODUCTION Neonatal encephalopathy per se is of course not just


due to hypoxia–ischemia. Seizures are one of the most
Birth represents a dramatic switch in life, from a fetus
obvious and common manifestations of encephalopathy
that relies on the placenta for respiratory exchange and
and can be used to help identify cases of encephalopathy.
nutrition, to an infant who must breathe for him- or her-
In a large contemporary cohort of infants (of whom 88%
self. While the majority of births are completely normal,
were term), the most common causes of neonatal seizures
complications that impair oxygen and glucose exchange
were HIE (38%), stroke (18%), and intracranial hemor-
can occur before or at birth at any gestational age. Acute rhage (11%) (Glass et al., 2016). Genetic epilepsy
brain injury around birth leading to disturbance of brain (6%) and metabolic problems such as hypoglycemia
function (i.e., neonatal encephalopathy) is associated (4%) were relatively uncommon. As well, hypoglycemia
with nearly half of all cases of cerebral palsy; of these, is a common complication of perinatal HIE and is asso-
the Western Australian Cerebral Palsy register found that ciated with worse outcomes (Basu et al., 2016). It is
approximately 15% of cases were born at term and had important to note that both neonatal encephalopathy
had acute encephalopathy at birth (Reid et al., 2016). and seizures are very common in preterm infants but
Hypoxic–ischemic encephalopathy (HIE) occurs in the the seizures are most often subclinical (Glass et al.,
developed world in approximately 2–3/1000 live births 2017). Stroke, hemorrhage, hypoglycemia, infection,
(Vannucci, 2000). Worldwide, HIE at birth and during and preterm brain injury are discussed in other chapters
the first 28 days of life represents the single greatest con-
in this volume (see Chapters 6, 7, 8, and 11). This chapter
tribution to overall disability worldwide and accounts for
will therefore focus on the pathophysiology and treat-
one-10th of all disability adjusted life years (Lee
ment of HIE at term and near-term.
et al., 2013).

*Correspondence to: Alistair J. Gunn, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019,
Auckland, New Zealand. Tel: +64-9-3737599x96763, E-mail: aj.gunn@auckland.ac.nz
218 A.J. GUNN AND M. THORESEN
In infants who have evidence of an acute, perinatal et al., 1975), and was associated with severe fetal acido-
event, the key link between exposure to asphyxia and sub- sis and hypoxia, hypotension, and tachycardia. Although
sequent neurodevelopmental impairment is the early onset there are no randomized controlled trials of temperature
of HIE. Newborns with mild encephalopathy are normal at control in labor, fever during labor has been indepen-
follow-up around 2 years of age, although they may be at dently associated with neonatal morbidity, including
risk of academic problems (Robertson et al., 1989; Odd death, neonatal seizures, encephalopathy and neonatal
et al., 2011). Nearly all infants with severe (Stage III) stroke in multiple studies, e.g., (Impey et al., 2008;
encephalopathy die or have severe disability, whereas Greenwell et al., 2012; Spain et al., 2015; Martinez-
only half of those with moderate (Stage II) HIE develop Biarge et al., 2016).
disability by 18 months to 2 years of age (Robertson Finally, approximately 10% of cases of moderate to
et al., 1989; Gluckman et al., 2005). However, studies severe neonatal encephalopathy have been reported to
from before the introduction of therapeutic hypothermia be associated with abnormal fetal heart rate recordings
suggest that even infants who do not develop neurologic before the start of labor, and so potentially may have been
impairment are at risk of more subtle neuromotor and associated with preceding fetal compromise (Westgate
academic problems (Robertson et al., 1989). et al., 1999; Jonsson et al., 2014). Chronic, moderate
hypoxia is not typical of labor; rather it is an antenatal
pattern. It may be associated with decreased fetal hemo-
Causes of pathologic hypoxia–ischemia globin (e.g., feto-maternal or feto-fetal hemorrhage),
placental insufficiency, and maternal causes such as pre-
A number of events may result in asphyxia around the
eclampsia. Naturally, there is potential for antenatal and
time of birth. Broadly, these may be grouped as repeated
intrapartum insults to be combined. Studies using mag-
hypoxia, acute catastrophic asphyxia, and chronic hyp-
netic resonance suggest that the great majority of infants
oxia (Westgate et al., 1999). Immediate, catastrophic
with acute HIE do not have established brain injury or
asphyxia contributes approximately 25% of cases of
atrophy, suggesting that injury still occurred relatively
HIE. This can be associated with cord prolapse, placental
soon before or during delivery (Cowan et al., 2003).
abruption, uterine rupture, vasa previa, fetal entrapment,
such as shoulder dystocia, and occasional cases of cord
entanglements and true knots in the cord. The impact
CHARACTERISTICS OF PERINATAL
of asphyxia during placental abruption may be greatly
ASPHYXIAL ENCEPHALOPATHY
potentiated by fetal blood loss leading to hypotension.
Next, repeated but relatively short periods of deep Perinatal HIE has a number of distinct characteristics
hypoxia are associated with approximately two-thirds that limit extrapolation from studies of the neonatal or
of cases of HIE at term (Westgate et al., 1999). As recently adult brain. First of all, the etiology of the insult is gen-
reviewed, this pattern is a direct function of the inherent erally global, affecting the whole fetus. Thus the fetal
intermittent “asphyxia” of labor (Lear et al., 2018). Intra- systemic and cardiovascular responses are critical to
partum uterine contractions impair gas exchange, leading understanding the pathogenesis of injury. Second, the
to transient fetal hypoxemia, hypercapnia, and metabolic insult is generally reversible, whether spontaneously or
acidemia (Bax and Nelson, 1993). This is mediated by therapeutically (e.g., delivery and resuscitation) and so
impaired uteroplacental perfusion during contractions, can be associated with an evolving pattern of cerebral
as shown by reduced uterine artery blood flow velocity dysfunction and delayed injury after the insult. Third,
(Fleischer et al., 1987; Janbu and Nesheim, 1987; Brar although the injury may be a single acute episode, it is
et al., 1988; Li et al., 2003) and reduced placental and commonly due to repeated insults. Fourth, many of the
intervillous perfusion (Ramsey, 1968; Sato et al., 2016; insults occur in the stable and warmer thermal environ-
Sinding et al., 2016). Frequent and long-lasting uterine ment of the uterus. Finally, the maturity of the brain
contractions, for example related to oxytocin augmenta- has a considerable effect on how neurons and glia
tion, will greatly reduce placental perfusion and so impair respond to asphyxia.
fetal oxygenation (Lear et al., 2018). It is now understood that the fetal response to
Further, maternal hyperthermia directly affects fetal asphyxia is not stereotypical, but rather depends upon
temperature and risk of neural injury. In pregnant both the nature of the insult and the condition of the fetus
baboons, raising the maternal temperature to between (Gunn and Bennet, 2009). In fact, it appears that the fetus
41 and 42°C increased uterine activity and increased is spectacularly good at defending itself against hypoxia,
the normal temperature gradient between fetus and and injury occurs only in a very narrow window between
mother from +0.47 to +0.75°C at 42°C (Morishima intact survival and death.
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 219
THE PATHOGENESIS OF CELL DEATH which causes neuronal depolarization (Hunter et al.,
2003). This regulated suppression of metabolic rate
What initiates neuronal injury? before energy stores are depleted during hypoxia or
At the most fundamental level, injury requires a period ischemia, termed adaptive hypometabolism, is actively
of insufficient delivery of oxygen and substrates such mediated through inhibitory neurotransmitters such as
as glucose (and other substrates in the fetus) such that gamma-aminobutyric acid (GABA) and adenosine
neurons and glia cannot maintain homeostasis. If the (Hunter et al., 2003), and protects the brain by delaying
neurons’ supply of high-energy metabolites such as the onset of cell depolarization. The duration of neuronal
adenosine triphosphate (ATP) is not able to be main- depolarization in turn determines the severity of injury
tained during hypoxia, then there is failure of the (Li et al., 2000).
energy-dependent mechanisms of intracellular homeo- Brain cells can use anaerobic metabolism to support
stasis, such as the Na+/K+ ATP dependent pump. Neu- production of high-energy metabolites for a limited
ronal depolarization then occurs, leading to sodium and time. This is extremely inefficient since whereas aero-
calcium entry into cells. This creates an osmotic and bic glycolysis produces 38 ATP with complete oxida-
electrochemical gradient that in turn favors further cat- tion of glucose, anaerobic glycolysis produces only
ion and water entry, leading to cell swelling (cytotoxic two ATP plus lactate, and so glucose reserves are rap-
edema). If sufficiently severe, this may lead to immedi- idly consumed, with a concomitant metabolic acidosis.
ate lysis (Johnston, 2005). The swollen neurons may Once fetal oxygen delivery has been restored, the fetus
still recover, at least temporarily, if the hypoxic is able to consume this circulating lactate. Many fetal
insult is reversed or the environment is manipulated. tissues, such as the heart, get a high proportion of their
Evidence suggests that several additional factors act substrate from sources other than glucose, particularly
to increase cell injury during and following depolariza- lactate (Fisher et al., 1982), and the brain can oxidize
tion. These include the extracellular accumulation of lactate when its concentration is elevated in fetal
excitatory amino acid neurotransmitters due to impair- sheep (Turbow et al., 1995) and very likely in human
ment of energy-dependent reuptake, which promotes newborn infants (Harris et al., 2015). However, as
further receptor-mediated cell swelling, excessive intra- lactate requires oxygen to be metabolized, this
cellular calcium entry, and the generation of oxygen strategy is only possible if there is sufficient time for
free radicals and inflammatory cytokines (Wassink re-oxygenation between intermittent episodes of
et al., 2014). hypoxia and is not possible during sustained, severe
Nevertheless, it is critical to appreciate that these fac- asphyxia.
tors appear to be injurious almost entirely in the presence
of hypoxic cell depolarization. For example, glutamate is Neural responses to severe hypoxia–ischemia
far more toxic during hypoxia (or mitochondrial dys- In contrast, under conditions where both oxygen and
function) than during normoxia (Deng et al., 2006). Con- substrate delivery are reduced, the options for the
versely, in vitro, hypoxia can still trigger cell death neuron are much more limited, since less oxygen
despite glutamate receptor blockade, through apoptotic and less glucose are available to support anaerobic
mechanisms (Niquet et al., 2006). metabolism. This may occur during pure ischemia
(reduced tissue blood flow, such as occurs in stroke)
and of immediate clinical relevance, during conditions
Neural adaptation to hypoxia
of hypoxia–ischemia, i.e., a combination of reduced
If blood oxygen content is reduced, but blood flow and oxygen content with reduced tissue blood flow. In
substrate concentrations are not impaired (i.e., moderate the fetus, hypoxia–ischemia commonly occurs due
hypoxia that did not impair fetal blood pressure or brain to hypoxic cardiac compromise, which leads to sec-
perfusion), neural adaptations complement the cardio- ondary hypotension and hypoperfusion. Under these
vascular adaptations to hypoxia to delay and often conditions, depletion of cerebral high-energy metabo-
completely avoid cell depolarization. First, the brain lites will occur much more rapidly and profoundly,
can reduce nonobligatory energy consumption, with a while at the same time there may actually be less
transient switch to a high-voltage low-frequency electro- acidosis, both because there is much less glucose avail-
encephalographic (EEG) state that requires less oxygen able to be metabolized to lactate and because the insult
consumption (Boddy et al., 1974; Blood et al., 2003). evolves more quickly.
Next, as hypoxia–ischemia becomes more severe, neuro- These concepts help to explain the consistent observa-
nal activity ceases completely at a threshold above that tion that most cerebral injury after acute perinatal insults
220 A.J. GUNN AND M. THORESEN
occurs in association with hypotension and consequent concentrations do increase in hypoxic fetuses, consistent
tissue hypoperfusion. In contrast, although asphyxial with previous observations in postnatal animals. This
brain injury by definition requires exposure to an anaer- appears to represent an intracellular compensatory mech-
obic environment, there is only a crude correlation anism for sustaining short-term mitochondrial oxygen
between the severity of systemic acidosis and the severity delivery in a critical organ with high oxygen consump-
of injury in any paradigm (Gunn and Bennet, 2009). tion (Guiang et al., 1993).
Clinical studies confirm a similar imprecise relationship. Thus the fetus is largely dependent on a steady supply
For example, in a single center cohort study (Vesoulis of oxygen, and consequently it has many adaptive fea-
et al., 2017), 412 of 27,028 infants had an arterial cord tures, some unique to the fetus, which help it to maximize
blood pH 7.10. Of these, 35 of 85 infants with arterial oxygen availability to its tissues even during moderate
cord blood pH <7.00 and 34 of 327 infants with pH hypoxia (Table 10.1). Thanks to these adaptations, the
between 7.00 and 7.10 developed HIE. Thus, screening fetus normally exists with a surplus of oxygen relative
infants with pH 7.1 for HIE was highly specific to its needs, which provides a significant margin of safety
(98.7%) but only moderately sensitive (74.2%). Criti- when oxygen delivery is impaired. These adaptive fea-
cally, over 25% of infants with moderate to severe HIE tures include: higher basal blood flow to organs, the
had milder acidosis than this threshold and so diagnosis higher affinity of fetal hemoglobin for oxygen, greater
of HIE was delayed. sensitivity of hemoglobin to acid promoting release to
Asphyxia is defined as the combination of impaired tissues, the amount of oxygen that can be extracted at typ-
respiratory gas exchange (i.e., hypoxia and hypercapnia) ical fetal oxygen tensions, the capacity to significantly
accompanied by the development of metabolic acidosis. reduce energy-consuming processes, greater anaerobic
When we think about the impact on the brain of clinical capacity in many tissues, and the capacity to redistribute
asphyxia, it will be critical to keep in mind that this def- blood flow toward essential organs away from the
inition tells us much about things that are relatively easily periphery.
measured (fetal blood gases and systemic acidosis) and Additional structural features of the fetal circulation
nothing about the fetal blood pressure and perfusion of further support these adaptive features including the
the brain, the key factors that most directly trigger brain systems of “shunts,” such as the ductus arteriosus, and
injury. preferential blood flow streaming in the inferior vena
cava to avoid intermixing of oxygenated blood from
SYSTEMIC AND CARDIOVASCULAR
ADAPTATIONS TO ASPHYXIA
Table 10.1
The systemic adaptations of the fetus to whole body
asphyxia are critical to outcome. Most of the experimen- Cardiovascular and cerebrovascular adaptations to
tal studies of the pathophysiology of fetal asphyxia have asphyxia
been performed in the chronically instrumented fetal ● The healthy fetus has considerable aerobic and anaerobic
sheep, in utero. The sheep is a highly precocial species, reserves to successfully adapt to transient or mild hypoxia
whose neural development broadly approximates that ● The fetal defenses against hypoxia depend on the type and
of term human infants around 0.8–0.85 of gestation severity of the insult, maturation, and fetal well-being
and of 27–30-week preterm infants at 0.7 of gestation ● During mild hypoxia the fetus switches to low-frequency,
(McIntosh et al., 1979). The reader should note that high-amplitude quiet sleep with reduced oxygen
the baseline heart rate of the fetal sheep is approxi- consumption
mately 20 beats per minute higher than that of the ● During moderate hypoxia, blood flow is maintained or
human fetus. increased to key organs such as the heart, brain, and adrenal
gland to enable normal brain metabolism. Blood flow is
redirected within the brain toward the structures important
Fetal adaptations and defense mechanisms
for autonomic function such as the brainstem
The fetus is highly adapted to intrauterine conditions, ● During severe asphyxia of rapid onset, the adaptations are
which include low partial pressures of oxygen and similar but more extreme. Blood flow to the brain as a whole
relatively limited supply of substrates compared with may not be increased but is typically maintained so long as
postnatal life. There are no material fetal reserves of fetal blood pressure is normal or raised
oxygen. Although tissue myoglobin could in theory act ● Progression of severe asphyxia results in failure of
as an oxygen store, in practice the fetus does not adaptation and progressive hypotension and hypoperfusion.
The severity of brain injury is consistently related to the
have appreciable tissue myoglobin levels except in
severity and duration of hypotension
the heart (Longo et al., 1973). Myocardial myoglobin
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 221
the placenta and deoxygenated blood in the fetal venous Table 10.2
system. These features ensure maximal oxygen delivery Acidosis and the pathogenesis of cell death.
to essential organs such as the brain and heart (Reuss
et al., 1981). During mild to moderate placental restric- ● There is no intrinsic, physiologic relationship between the
tion, fetal oxygen consumption can be maintained at nor- amount of systemic anaerobic metabolism (as reflected by
mal levels until uteroplacental blood flow falls below metabolic acidosis) and the development of neuronal injury.
The crude clinical correlation between the two simply
50% (Wilkening and Meschia, 1983), with normal
reflects that hypoxic–ischemic damage occurs under
removal of waste products of metabolism, mainly carbon
anaerobic conditions
dioxide, water, and heat (Gilbert et al., 1985; Giussani ● Systemic acidosis during asphyxia is primarily related
et al., 1993). to peripheral vasoconstriction, which acts to redistribute
Cerebral oxygen consumption is even more protected blood flow to essential organs. Even when a proportion
and is little changed even if arterial oxygen content falls of whole body anaerobic metabolism is central, so long
as low as 1.5 mM (compared with about 4 mM in the nor- as sufficient glucose is available to neurons to support
mal fetus), thanks to compensating increases in both basal energy metabolism, injury will not occur. Thus
cerebral blood flow (CBF) and oxygen extraction profound arterial metabolic acidosis may still
(Peeters et al., 1979). Nitric oxide (NO) and adenosine accompany successful protection of the brain, with a
have been shown to play a role in mediating the local normal outcome
increase in CBF during hypoxia (Green et al., 1996; ● During very prolonged exposure to intermittent
Blood et al., 2003). asphyxia, with ongoing lactic acidosis, very severe
acidosis may compromise fetal adaptation, including
cardiac contractility and blood flow redistribution,
promoting hypotension and thus causing cerebral
Etiology of asphyxia hypoxia–ischemia
ACUTE PROFOUND ASPHYXIA ● During acute severe insults that lead to
hypotension-impaired tissue perfusion, less glucose is
Studies of asphyxia by definition involve both hypoxia delivered to neurons and so less lactate is produced.
and hypercapnia with metabolic acidosis (e.g., see Thus injury may occur despite relatively modest levels
Table 10.2). It is important to appreciate that such stud- of acidosis
ies of asphyxia typically use much greater depth of ● Acute or chronic insults, where preexisting reduced
hypoxia than is possible using maternal inhalational fetal metabolic reserve is followed by sudden onset of
hypoxia. Further, asphyxia can be induced relatively deeper hypoxia, have the potential to lead to injury after
abruptly, limiting the time available for adaptation. relatively short periods of acute asphyxia and moderate
Complete clamping of the uterine artery or umbilical acidosis
cord leads to a rapid reduction of fetal oxygenation
over the first minute (Bennet et al., 1998). In contrast,
Uterine contractions and brief repeated asphyxia
gradual partial occlusion induces a slow fetal metabolic
deterioration without the initial fetal cardiovascular Although the fetus can be exposed to a wide range of
responses of bradycardia and hypertension, reflecting insults during labor, the key, distinctive characteristic
the greater time available for fetal adaptation and lesser of labor is brief, intermittent, repetitive episodes of
degree of hypoxia (de Haan et al., 1993). During asphyxia, which are almost entirely related to uterine
profound asphyxia, corresponding with a severe reduc- contractions (Westgate et al., 2007). During normal
tion of uterine blood flow to 25% or less and a fetal labor there is intermittent reduction of placental gas
arterial oxygen content of less than 1 mmol/L, the exchange that is associated with a consistent fall in pH
fetal cardiovascular responses are substantially dif- and oxygen tension, and a rise in carbon dioxide and base
ferent from those during moderate hypoxia or asphyxia deficit in normal, uncomplicated labor (Wiberg et al.,
(Parer, 1998). 2006). Typically, the second stage of normal labor will
Three broad phases of the cardiovascular responses of be the time of greatest hypoxic stress for the fetus and
the fetus to such severe events can be distinguished: the is accompanied by a more rapid decline in pH and trans-
initial, rapid chemoreflex-mediated adaptations; sus- cutaneous oxygen tension (Modanlou et al., 1974;
tained adaptation that is supported by circulating, Peebles et al., 1994) and rise in transcutaneous carbon
humoral factors; and the final period of progressive hyp- dioxide tension (Katz et al., 1987). Thus, in a technical
oxic decompensation ultimately terminated by profound sense, essentially all fetuses may be said to be exposed
systemic hypotension and cerebral hypoperfusion, as to “asphyxia” during labor. Fortunately, it is usually
recently reviewed in depth (Bennet, 2017). mild and well tolerated by the fetus. Unfortunately,
222 A.J. GUNN AND M. THORESEN
both the lay public and many clinicians associate the term understood since reduced perfusion will reduce supply of
asphyxia with the development of severe metabolic glucose for anaerobic metabolism, compounding the
acidosis, postasphyxial encephalopathy, and other end- reduction in oxygen delivery and concentration. The
organ damage or death. In our haste to avoid using the real-life importance of hypotension is supported by both
term, the normal nature of labor and its effects on the the correlation of injury with arterial blood pressure
fetus are often not fully appreciated. across multiple paradigms, and by the common patterns
Most fetuses enter labor with a large reserve of of neural damage.
placental capacity that helps accommodate the repeated During severe asphyxia, mean arterial blood pressure
brief reductions in oxygen supply during contractions. (MAP) initially rises due to intense peripheral vasocon-
Contraction strength, frequency, and duration are the striction (Bennet, 2017). At this time cerebral blood flow
key factors that determine the rate at which fetal asphyxia is maintained. If asphyxia is continued, MAP eventually
develops during labor. Critically, the proportion of falls, typically after 7 min in term fetal sheep, as a func-
time the uterus spends at resting tone compared with tion of impaired cardiac contractility and failure of
contracting tone will determine the extent to which fetal peripheral redistribution. Once MAP falls below base-
gas exchange can be restored between contractions line, carotid blood flow falls in parallel, consistent with
(Huch et al., 1977). Thus, any intervention that increases the known, relatively narrow, cerebral autoregulatory
the frequency and/or duration of uterine contractions range in the fetus (Parer, 1998).
clearly places the fetus at increased risk of compromise. In the near-term fetus neural injury has been com-
For example, studies using near-infrared spectroscopy monly reported in areas such as the parasagittal cortex,
(NIRS) show a progressive fall in cerebral oxygen satu- the dorsal horn of the hippocampus, and the cerebellar
ration when contractions occur more frequently than neocortex after a range of insults including pure ische-
every 2.3 min (Peebles et al., 1994). The effects of mia, prolonged single complete umbilical cord occlu-
repeated hypoxia may be amplified in vulnerable fetuses, sion, and prolonged partial asphyxia and repeated brief
for example in those with preexisting placental insuffi- cord occlusion (e.g., as illustrated in Fig. 10.1, bottom
ciency (Westgate et al., 2005; Wassink et al., 2013) or panel) (Gunn et al., 1992, 1997; Mallard et al., 1992;
fetal inflammation leading to sensitization of the brain de Haan et al., 1993, 1997a). These areas are watershed
to hypoxia–ischemia (Eklind et al., 2005; Osredkar zones within the borders between major cerebral arteries,
et al., 2014). Conversely, even a normal fetus with nor- where perfusion pressure is least. In both adults and chil-
mal placental function may be unable to fully adapt to dren lesions in these areas are typically seen after sys-
tonic contractions or uterine hyperstimulation related temic hypotension (Torvik, 1984).
to oxytocin infusion used for induction or augmentation There are some data suggesting that mild white or
or prostaglandin preparations for induction of labor gray matter injury can occur even in the absence of sig-
(Winkler and Rath, 1999). nificant hypotension (de Haan et al., 1993; Ikeda et al.,
1998), particularly when hypoxia is very prolonged
(Rees et al., 1998). There may have been local or relative
PATHOPHYSIOLOGIC DETERMINANTS
hypoperfusion in these studies even if overall perfusion
OF ASPHYXIAL BRAIN INJURY
was maintained. Nevertheless, there is a strong correla-
Recent studies using well-defined experimental para- tion between either the depth or duration of hypotension
digms of asphyxia in the near-term fetal sheep have and the amount of neuronal loss within individual studies
explored the relationship between the distribution of neu- of acute asphyxia (Gunn et al., 1992; de Haan et al.,
ronal damage and the type of insult. These studies sug- 1997a, b; Ikeda et al., 1998). This is also seen between
gest that, while local cerebral hypoperfusion due to similar asphyxial paradigms causing severe fetal acidosis
hypotension is required to cause injury, a number of fac- that have been manipulated to either cause fetal hypoten-
tors including the pattern of repetition of insults as well as sion (Gunn et al., 1992) or not (de Haan et al., 1993). In
fetal factors such as maturity, preexisting metabolic state, fetal lambs exposed to prolonged severe partial asphyxia,
and cerebral temperature markedly alter the impact of the as judged by the degree of metabolic compromise, neu-
insult on the brain (Gunn and Bennet, 2009). ronal loss occurred only in those in whom one or more
episodes of acute hypotension occurred (Gunn et al.,
1992). In a study where an equally severe insult was
Hypotension and the watershed distribution
induced gradually, but titrated to maintain normal or ele-
of neuronal loss
vated blood pressure throughout the insult, no neuronal
The development of hypotension is highly associated loss was seen except in the cerebellum (de Haan
with neural injury during acute asphyxia. This is readily et al., 1993).
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 223
led to a greater proportion of striatal injury relative to cor-
tical neuronal loss (Mallard et al., 1995). Intriguingly,
significant striatal involvement was also seen after pro-
longed partial asphyxia in which distinct episodes of bra-
dycardia and hypotension occurred (Gunn et al., 1992).
Given that both parasagittal and basal ganglia predomi-
nant patterns of injury are seen on early MRI scans in
term infants with encephalopathy (Miller et al., 2005),
intermittent severe insults are likely to be clinically
important.
The striatum is within the territory of the middle cere-
bral artery and not a watershed zone. Thus it is likely that
the pathogenesis of striatal involvement in the near-term
fetus is related to the precise timing of the relatively pro-
longed episodes of asphyxia and not to more severe local
hypoperfusion. Speculatively, the apparent vulnerability
of striatal medium-sized neurons to this type of insult
may be related to a greater release of glutamate into
the extracellular space after repeated insults compared
with a single insult of the same cumulative duration.
Consistent with this, immunohistochemical techniques
have shown that inhibitory striatal neurons were primar-
ily affected (Mallard et al., 1995).

Fig. 10.1. The distribution of neuronal loss assessed after Cerebral injury: An evolving process
3 days recovery from two different patterns of prenatal
The key concept to emerge from both experimental and
asphyxia in near-term fetal sheep. The left panel shows the
clinical studies is that brain cell death does not necessar-
effects of brief (1 or 2 min) cord occlusions repeated at fre-
quencies consistent with established labor. Occlusions were ily occur during hypoxia or ischemia, but rather they may
terminated after a variable time, when the fetal blood pressure precipitate a cascade of biochemical processes leading to
fell below 20 mmHg for two successive occlusions. This insult delayed cell death hours or even days afterwards (the sec-
led to damage in the watershed regions of the parasagittal cor- ondary phase, Fig. 10.2). It is now well established in
tex and cerebellum (de Haan et al., 1997a). The right panel term infants and animals that there can be considerable
shows the effect of 5-min episodes of cord occlusion, repeated cell survival and recovery of oxidative metabolism after
four times, at intervals of 30 min (de Haan et al., 1997b). This severe HI in a so-called latent phase, followed by pro-
paradigm is associated with selective neuronal loss in the puta- gressive secondary failure of oxidative metabolism and
men and caudate nucleus, which are nuclei of the striatum. The evolution of cell death over hours to days (Wyatt et al.,
cornu ammonis and the dentate gyrus are regions of the hippo-
1989; Lorek et al., 1994).
campus. Mean  SD.
During the immediate period of hypoxia–ischemia
(which may be termed the primary phase of the insult)
high-energy metabolites are depleted, with progres-
The pattern of injury: Repeated insults
sive depolarization of cells, severe cytotoxic edema
of moderate duration
(cell swelling, e.g., see Fig. 10.3) (Gunn et al.,
One apparent exception to a general tendency to a water- 1997) and extracellular accumulation of excitatory
shed distribution after global asphyxial insults in the amino acids due to failure of reuptake by astroglia
near-term fetus is the selective neuronal loss in striatal and excessive depolarization mediated release (Tan
nuclei (putamen and caudate nucleus, Fig. 10.1, top et al., 1996). Although neurons may die during a
panel), which develops when relatively prolonged sufficiently prolonged period of ischemia or asphyxia,
periods of asphyxia or ischemia are repeated (Mallard many neurons initially recover, at least partially, from
et al., 1995; de Haan et al., 1997a). Whereas 30 min of the insult in a so-called latent phase, only to die many
continuous cerebral ischemia was associated with pre- hours or even days later (secondary or delayed cell
dominantly parasagittal cortical neuronal loss, with only death). Studies using magnetic resonance spectro-
moderate striatal injury, 3  10 min periods of ischemia scopy showed that many infants with evidence of
224 A.J. GUNN AND M. THORESEN
Phases of cerebral injury
Hypothermia

Insult Latent Secondary Tertiary


Hypoxic From 1 to 6 h From ~6 h to 3 d Months
depolarization Recovery of oxidative Mitochondrial failure Remodeling
EAAs metabolism vs residual
Seizures/EAAs Chronic
mitochondrial injury
OFRs/NO Cytotoxic edema inflammation
Programmed cell death
Calcium entry Final cell death Astrogliosis
2° inflammation
Cell lysis
Receptor hyperactivity

Reperfusion
Fig. 10.2. Flowchart illustrating the relationship between the mechanisms active in the pathophysiologically defined phases of
cerebral injury after a severe reversible hypoxic–ischemic insult. During the immediate reperfusion period, lasting approximately
30 min, cellular energy metabolism is restored, with resolution of the acute hypoxic depolarization and cell swelling. This is
followed by a latent phase, with near-normal oxidative cerebral energy metabolism, as shown by magnetic resonance spectros-
copy, but depressed electroencephalogram (EEG) activity, and often a delayed period of reduced CBF. The latent phase is believed
to be associated with induction of the intracellular components of the apoptotic cascade. This may be followed by secondary dete-
rioration with delayed seizures and cytotoxic edema, extracellular accumulation of potential cytotoxins (such as the excitatory
neurotransmitters), and, 4–15 h after the asphyxia, failure of oxidative metabolism and damage. The changes in the secondary
phase may take 3 days or more to resolve.

moderate to severe asphyxia show initial, transient failure and loss of ability to produce high-energy phos-
recovery of cerebral oxidative metabolism after birth, phates (Tsuji et al., 1995). Continuous, noninvasive mea-
followed by secondary deterioration as shown by surements with NIRS demonstrate that after severe
delayed cerebral energy failure from 6 to 15 h after asphyxia in fetal sheep there is initial recovery of CytOx
birth (Azzopardi et al., 1989). The severity of the values, followed by a progressive fall, starting from
secondary deterioration is closely correlated with neu- approximately 3–4 h and continuing until approximately
rodevelopmental outcome at 1 and 4 years of age 48–72 h after asphyxia (Bennet et al., 2006). Delayed
(Roth et al., 1997), and infants with encephalopathy loss of mitochondrial activity was associated with a
who do not show initial recovery of cerebral oxidative marked increase in relative intracerebral oxygenation
metabolism have extremely poor outcomes (Azzopardi consistent with impaired ability to use oxygen (Bennet
et al., 1989). et al., 2006).
An identical pattern of initial recovery of cerebral oxi- Characteristic pathophysiologic changes may be
dative metabolism followed by delayed (secondary) distinguished during this critical latent phase compared
energy failure is also seen after hypoxia–ischemia in with the secondary phase (Figs. 10.2–10.4). After resto-
the piglet, rat, and fetal sheep and is closely correlated ration of circulation and oxygenation, the initial hypoxia-
to the severity of neuronal injury (Lorek et al., 1994; induced impairments of cerebral oxidative metabolism,
Blumberg et al., 1997; Bennet et al., 2006). The timing cytotoxic edema and accumulation of excitatory
of energy failure after hypoxia–ischemia is tightly amino acids resolve over approximately 30–60 min
coupled with the appearance of histologic brain damage (Tan et al., 1996; Gunn et al., 1997). Despite normaliza-
(Vannucci et al., 2004), implying that it is primarily a tion of oxidative cerebral energy metabolism and mito-
function of evolving cell death. chondrial activity (Bennet et al., 2006), mean
It is this delay before secondary deterioration that electroencephalogram (EEG) activity remains depressed
offered the tantalizing possibility that therapeutic inter- (Keogh et al., 2012), while CBF initially recovers, fol-
vention after hypoxia–ischemia might be possible. More lowed by a transient secondary fall (Gunn et al., 1997;
recent studies have provided a detailed time course of the Bennet et al., 2006). During the subsequent secondary
evolution of injury. Because oxidative synthesis of ATP deterioration starting many hours later (typically approx-
is mediated through the mitochondrial electron transport imately 6–15 h after moderate to severe hypoxia–
chain, loss of the terminal electron acceptor, cytochrome ischemia) delayed seizures develop and then continue
oxidase (CytOx), is a close surrogate for mitochondrial for several days (Gunn et al., 1997; Bennet et al., 2006),
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 225
ultimately, cell death (Gunn et al., 1997). In contrast,
secondary edema and seizures are not seen after milder
insults that do not cause cortical injury (Williams
et al., 1991).
It is important to appreciate that the severity of
HI markedly affects the speed of evolution of cell
death. For example, in 21-day-old rat pups (broadly
equivalent to late infancy in humans), 15 min of hyp-
oxia–ischemia was associated with selective neuronal
death in an apoptotic morphology from 3 days after
hypoxia (Beilharz et al., 1995). By contrast, after
60 min of hypoxia–ischemia, some cells showed DNA
degradation by 10 h after hypoxia, and widespread cor-
tical necrosis developed after 24 h. Moreover, as well
as evolving over time, cell death spreads outward from
the most severely affected regions toward less severely
affected regions (Thornton et al., 1998). In piglets
exposed to transient HI, the cerebral apparent diffusion
coefficients (ADCs) normalized almost completely by
2 h after resuscitation, followed in the majority of ani-
mals by a fall in ADC beginning in the parasagittal cortex
and then spreading through the brain. This pattern likely
reflects both severity of injury and active mechanisms
including opening of astrocytic connexin hemichannels
on the cell surface, which can facilitate waves of spread-
ing depression that can trigger cell death in less injured
tissues (Davidson et al., 2015a).
The secondary phase resolves over 3 days after
severe hypoxia–ischemia into a tertiary phase of ongoing
Fig. 10.3. Changes in fetal extradural temperature (top panel), injury, involving repair and reorganization that may last
cortical impedance (middle) and electroencephalogram (EEG) weeks to months and even years (Hagberg et al., 2015;
power (bottom), after 30 min of global cerebral ischemia in the Bennet et al., 2018). Chronic inflammation and epige-
near-term fetal sheep. Impedance is a measure of cell swelling
netic changes lasting for weeks to months after injury
in the parietal cortex. Fetuses received either sham-ischemia plus
sham cooling (), or ischemia with sham cooling (●) or cerebral
may impair optimal neurorepair and functional recovery
hypothermia ( ) started 3 h after reperfusion and continued until (Fleiss and Gressens, 2012; Bennet et al., 2018; Galinsky
72 h (shown by blue bar). The end of cerebral ischemia is shown et al., 2018). Key neuroprotection strategies in this phase
by vertical arrow at time 0. The hypothermia group shows include alleviating chronic local inflammation and
complete suppression of the secondary rise in impedance stimulation of endogenous factors that can support pro-
(i.e., it prevented delayed cell swelling), and substantially liferation, migration, and maturation of glia and neurons
better recovery of EEG intensity after resolution of delayed (Hagberg et al., 2015; Bennet et al., 2018). Treatments
seizures. Secondary hypoperfusion is extended by hypothermia such as stem cell therapy potentially may be beneficial
but resolves spontaneously despite continued cooling for 72 h in this phase, given their multimodal effects in
(not shown). Data derived from Davidson, J.O., Draghi, V., reducing neural inflammation and promoting release of
Whitham, S., et al., 2018b. How long is sufficient for optimal
trophic factors (Fleiss et al., 2014; van den Heuij et al.,
neuroprotection with cerebral cooling after ischemia in fetal
sheep? J Cereb Blood Flow Metab 38, 1047–1059; Davidson,
2017; Bennet et al., 2018). Further, there is a clear
J.O., Wassink, G., Yuill, C.A., et al., 2015b. How long is too role for postnatal neurorehabilitation for optimizing
long for cerebral cooling after ischemia in fetal sheep? J Cereb development of the neural network (Pitcher et al.,
Blood Flow Metab 35, 751–758. Mean  SEM, *P < 0.05. 2009; Maitre, 2015). Importantly, as part of the chal-
lenges to improving overall outcomes after HIE, early
accompanied by secondary cytotoxic edema (cell and accurate diagnosis of movement disorders such as
swelling) (Gunn et al., 1997), accumulation of excito- cerebral palsy will make a significant difference in
toxins (Tan et al., 1996), failure of cerebral mitochondrial providing the right neurorehabilitation treatment, at the
activity (Lorek et al., 1994; Bennet et al., 2006), and, right time (Novak et al., 2017).
226 A.J. GUNN AND M. THORESEN
These concepts, that an acute, global insult can mortality or major neurodevelopmental disability by
trigger evolving injury and that characteristic events 18 months of age (relative risk (RR) 0.75 (95% confi-
are seen at different times after the insult, are central dence interval (CI) 0.68–0.83)). Cooling reduced mortal-
to understanding the causes and treatment of neonatal ity (RR 0.75 (95% CI 0.64–0.88), 11 studies, 1468
encephalopathy. infants), with reduced neurodevelopmental disability in
survivors (typical RR 0.77 (95% CI 0.63–0.94), 8 stud-
ies, 917 infants).
THERAPEUTIC HYPOTHERMIA
Long-term follow-up of these studies is ongoing; the
Compelling experimental evidence from multiple pre- available evidence suggests a similar improvement in
clinical paradigms (Gunn and Thoresen, 2006) led to outcomes in middle childhood after mild induced hypo-
multiple randomized controlled trials (RCTs) of mild thermia for HIE (Guillet et al., 2012; Shankaran et al.,
induced hypothermia for moderate to severe neonatal 2012; Azzopardi et al., 2014). For example, the Total
HIE. The first large animal neonatal study, for example, Body Hypothermia for Neonatal Encephalopathy Trial
showed that mild hypothermia, reducing core tempera- has shown that significantly more children in the mild
ture from 38.5 to 35°C, started quickly after hypoxia– hypothermia group survived with an IQ score of 85 or
ischemia in newborn piglets and continued for 12 h, more compared to the control group (52% vs 39%, RR
prevented the delayed loss of high-energy phosphates 1.31, P ¼ 0.04), and that more children in the hypother-
that developed in normothermic animals. This protection mia group survived without neurologic abnormalities
persisted to 72 h after hypoxia–ischemia (Fig. 10.4) than in the control group (45% vs 28%, RR 1.60, CI:
(Thoresen et al., 1995). 1.15, 2.22) (Azzopardi et al., 2014). Further, there was
A systematic meta-analysis of 11 RCTs of either a significant reduction in the risk of cerebral palsy
selective head cooling or whole body cooling initiated (21% vs 36%, P ¼ 0.03) and of moderate or severe dis-
within 6 h of birth, involving 1505 term and late preterm ability (22% vs 37%, P ¼ 0.03). Moreover, recent cohort
infants with moderate/severe HIE, found highly consis- studies of infants cooled for HIE showed a lower inci-
tent beneficial effects after hypothermia (Jacobs et al., dence of epilepsy at 2 years of age compared with the
2013). Mild hypothermia was associated with reduced cooling trials (Liu et al., 2017) as well as reduced severity

Fig. 10.4. Mild hypothermia prevents secondary energy failure. One-day-old anesthetized term newborn pigs underwent bilateral
carotid occlusion with global mild hypoxia until ATP levels, as analyzed by spectroscopy, had fallen to at least 30% of normal
values (Lorek et al., 1994; Thoresen et al., 1995). Proton and phosphorous spectroscopy was undertaken at preset time-points for up
to 72 h. The pigs were ventilated and fed intravenously inside the bore of the magnet during this time. There were three groups,
anesthetized sham controls without an insult (triangles, dotted line), hypoxia–ischemia followed by normothermic recovery for
48 h (open circles, dashed line), and hypoxia–ischemia followed by hypothermia for 12 h (core temperature reduced by 3.5°C)
starting after reoxygenation and reperfusion, then normothermic recovery after hypothermia for a total of 64 h (black diamonds,
solid line); 12 h of hypothermia ameliorated the secondary energy failure seen during normothermic recovery.
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 227
of cerebral palsy (Jary et al., 2015). The reader should and then progressively fell (Bennet et al., 2006). Imme-
note that it is not possible to exclude the possibility that diate initiation of hypothermia has been shown to be
infants with less severe HIE were recruited once thera- protective in many studies (Wassink et al., 2018). For
peutic hypothermia became standard care. Nevertheless, example, in anesthetized piglets exposed to either hyp-
these studies used the same entry criteria for hypothermia oxia with bilateral carotid ligation or to hypoxia with
as the original cooling trials, including amplitude inte- hypotension, either 12 h of mild whole body hypother-
grated EEG monitoring. mia (35°C) or 24 h of head cooling with mild systemic
hypothermia started immediately after hypoxia pre-
IS IT POSSIBLE TO FURTHER OPTIMIZE vented delayed energy failure, reduced neuronal loss,
THERAPEUTIC HYPOTHERMIA? and suppressed posthypoxic seizures (Thoresen et al.,
1995; Tooley et al., 2003).
Despite significantly reducing death and disability, cur-
A sufficiently prolonged duration of hypothermia
rent protocols for therapeutic hypothermia are only par-
may be protective despite delayed initiation. In the
tially neuroprotective, with a number needed to treat of 7
near-term fetal sheep, moderate hypothermia induced
((95% CI 5–10), 8 studies, 1344 infants) (Jacobs et al.,
90 min after reperfusion from a severe episode of cerebral
2013). As reviewed, the experimental efficacy of hypo-
ischemia, in the early latent phase, and continued until
thermia is highly dependent on the timing of initiation,
72 h after ischemia prevented secondary cytotoxic edema
depth, and duration of cooling (Wassink et al., 2018).
and improved electroencephalographic recovery (e.g.,
Thus, potentially, it may be possible to further optimize
see Fig. 10.3) (Gunn et al., 1997). There was a concom-
regimens for therapeutic hypothermia.
itant substantial reduction in cortical infarction and
improvement in neuronal loss scores in all regions
At the same time, it is important to appreciate that
(Fig. 10.5). Similar neuroprotective effects were seen
there is some evidence that outcomes may have
when treatment was delayed until 3 h after the end of
improved further now that therapeutic hypothermia is
ischemia (Davidson et al., 2018b). However, when the
routine care for moderate to severe HIE. In a recent ran-
start of hypothermia was delayed until just before the
domized controlled trial of cooling strategies, the rate of
onset of secondary seizures (5.5 h after reperfusion) only
death or disability at 18 months of age in infants treated
partial neuroprotection was seen (Gunn et al., 1998).
with cooling to 33.5°C for 72 h was 29.3% (Shankaran
With further delay until after seizures were established
et al., 2017) compared with 44% in infants receiving
(8.5 h after reperfusion), there was no electrophysiologic
the same cooling protocol and recruited using the same
or overall histologic protection with cooling (Gunn
criteria in a previous trial (Shankaran et al., 2005). The
et al., 1999).
factors behind this apparent improvement are not known.
Similar results have been reported in a wide range
Speculatively, it might be related to recruiting infants
of species and paradigms. For example, in unanesthe-
with less severe HIE as shown by less severe Apgar
tized 21-day-old rat pups mild hypothermia (2–3°C
scores (Thoresen et al., 2013), but it might also partly
decrease in brain temperature) for 72 h after hypoxia–
be related to earlier initiation of cooling, with increasing
ischemia prevented cortical infarction, whereas cooling
use of passive cooling while infants are assessed for
delayed until 6 h after the insult had an intermediate,
active cooling (Lemyre et al., 2017). A generic clinical
nonsignificant effect (Sirimanne et al., 1996). More
hypothermia protocol is shown in Supplementary
recently, in 7-day-old rat pups, hypothermia was found
Material in the online version at https://doi.org/
to be notably protective after a “moderate” duration of
10.1016/B978-0-444-64029-1.00010-2.
hypoxia–ischemia (90 min) (Sabir et al., 2012), such
that immediate induction of hypothermia after moderate
The timing of initiation of hypothermia
hypoxia–ischemia significantly reduced the area of
There is highly consistent evidence that hypothermia cortical infarction (P < 0.05), with a linear loss of
must be started as early as possible within the latent effect with greater delay in starting cooling, up to 6 h
phase for optimal benefit. This concept is highly consis- of delay (Fig. 10.6A). This window of opportunity is
tent with the finding of progressive mitochondrial failure strikingly similar to the studies in fetal sheep noted
during the latent phase demonstrated by magnetic reso- previously (Fig. 10.5). By contrast, even immediate
nance spectroscopy after moderate to severe hypoxia– hypothermia did not improve outcomes after very pro-
ischemia in human infants (Azzopardi et al., 1989; longed hypoxia–ischemia (150 min, Fig. 10.6B). This
Roth et al., 1997) and piglets (Thoresen et al., 1995; illustrates the critical point that the window of opportu-
Fig. 10.4). Similarly, in preterm fetal sheep, levels of nity for therapeutic hypothermia is critically dependent
oxidized mitochondrial cytochrome oxidase levels were on the severity of the primary period of hypoxia–
normal to elevated in the first 3 h after severe asphyxia ischemia.
228 A.J. GUNN AND M. THORESEN

Fig. 10.5. Time-dependent neuronal rescue with delayed cerebral hypothermia. Effect of early (90 min) or delayed (5.5 h) or late
(8.5 h) cerebral cooling after global cerebral ischemia for 30 min on microscopically assessed neuronal loss in different brain
regions at 5 days after ischemia in near-term fetal sheep. A significant reduction (P < 0.001) in neuronal loss was seen in all regions
in fetuses treated with early cerebral cooling compared with sham cooled fetuses, demonstrating that delayed neuronal death can be
prevented. Cooling delayed until 5.5 h after ischemia, just before the start of the secondary phase, was associated with partial pro-
tection. Note: In the early hypothermia group, only fetuses in which the extradural temperature was maintained below 34°C for the
first 12 h are shown (Gunn et al., 1997, 1998, 1999). Mean  SEM.

Clinical evidence supports the importance of initiat- Colbourne et al., 1998). By contrast, in 7-day-old rat pups,
ing hypothermia as early as possible. In a cohort study, cooled to 33.5, 32, 30, 26, or 18°C for 5 h after hypoxia–
infants who were treated within 3 h after birth had signif- ischemia, there was no additional protection with temper-
icantly better motor outcomes than those treated after 3 h atures below 33.5°C (Wood et al., 2016). Similarly, in
(Thoresen et al., 2013). However, in a controlled trial, neonatal piglets, whole body hypothermia with a reduc-
hypothermia was only started in 12% of infants within tion in body temperature of either 3.5°C or 5°C was
4 h of birth; at that time many infants were already show- associated with significant (and highly similar) overall
ing electrographic seizures (Gluckman et al., 2005). This neuroprotection, whereas a reduction of 8°C was
is earlier onset of seizures than typically seen in preclin- associated with loss of protection after global cerebral
ical studies (Williams et al., 1990), consistent with clin- ischemia (Alonso-Alconada et al., 2015). Finally, a large
ical evidence that many hypoxic–ischemic insults evolve randomized controlled clinical trial has confirmed that
over time before birth and so the timing of the insult is cooling infants with moderate to severe HIE to 32°C
often not clearly known (Westgate et al., 1999). More- instead of 33.5°C did not further reduce death or moderate
over, accurate staging of encephalopathy can be difficult or severe disability at 18 months of age (Shankaran et al.,
in the critical latent phase of injury, and so diagnosis may 2017). These consistent clinical and preclinical findings
be delayed until after the onset of seizure activity, by suggest that there is a relatively broad range of tempera-
which time the efficacy of hypothermia is greatly tures beneficial for the brain after hypoxia–ischemia, and
reduced. that, reassuringly, it should not be generally necessary to
reduce core temperatures by more than 3.5°C.
How deep is too deep?
If some is good, is more better?
The critical depth of hypothermia required for protection
may be affected by multiple factors, including the delay There is now compelling animal evidence that continu-
before initiation and the severity and nature of the insult. ing cooling for approximately 72 h provides optimal neu-
There is some evidence from studies of moderate to roprotection (Davidson et al., 2015b, 2018b). Broadly,
severe ischemia that when cooling was delayed until and critically for clinical practice, the greater the delay
6 h, in both adult rodents and fetal sheep, greater func- before starting cooling, or the more severe the insult,
tional and histological neuroprotection was seen with a the greater the duration of cooling required for protec-
5°C reduction than with 3°C (Gunn et al., 1997; tion. For example, in adult gerbils when the delay before
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 229

Fig. 10.6. Effect of delay before initiating hypothermia and severity of hypoxia–ischemia on hypothermic neuroprotection. This
figure shows area loss in the ligated hemisphere after 1 week recovery in 7-day-old rats undergoing unilateral carotid ligation
followed by global inhalational hypoxia for defined times (Sabir et al., 2012). Five hours of hypothermia was initiated immediately,
or 3, 6, or 12 h after HI. Panel (A, top) shows time-dependent neuroprotection with postinsult hypothermia after a moderate period
of HI (8% O2 for 90 min with the environmental temperature maintained at 36°C), such that the sooner hypothermia was started, the
greater the reduction in area of infarction. Panel (B) shows that when the rat pups were exposed to a severe period of HI (8% O2 for
150 min with the environmental temperature maintained at 37°C), there was increased mortality and no neuroprotection from hypo-
thermia. Indeed, hypothermia started 12 h after the insult was associated with increased loss of brain area.

initiating a 24-h period of cooling was increased from 1 until 48 h, was partially protective, it was substantially
to 4 h, neuroprotection in the CA1 region of the hippo- less effective for both recovery of EEG power and neu-
campus after 6 months recovery fell from 70% to 12% ronal survival than cooling for 72 h (Davidson
(Colbourne and Corbett, 1995). Subsequent studies dem- et al., 2018b).
onstrated that protection could be restored by extending Given this compelling evidence that hypothermia must
the duration of moderate (32–34°C) hypothermia to 48 h be continued for at least 72 h in large animals and humans,
or more, even when the start of cooling was delayed until there has been interest in whether further prolonging the
6 h after reperfusion (Colbourne et al., 2000). Similarly, duration of hypothermia may be associated with greater
in the near-term fetal sheep, cooling started after 5.5 h benefit. However, in the term-equivalent fetal sheep when
and continued until 72 h was still partially protective delayed hypothermia starting 3 h after ischemia was pro-
(Gunn et al., 1998) (Fig. 10.5). Finally, in the same par- longed from 3 to 5 days, there was no further improvement
adigm, although delayed cooling, from 3 h after ischemia in electrophysiologic recovery or neuronal survival or
230 A.J. GUNN AND M. THORESEN
reduction in cortical microglial induction (Davidson et al., Should we cool infants with “mild” HIE?
2015b). Indeed, extended cooling was associated with a
The large RCTs of therapeutic hypothermia excluded
small reduction in neuronal survival in the parasagittal cor-
infants who had “mild” HIE in the first 6 h of life in order
tex and the dentate gyrus. Consistent with these experi-
to increase the rates of unfavorable outcome, and so the
mental data, a recent clinical trial of prolonged duration
potential benefit of treating these infants with therapeutic
and increased depth of therapeutic hypothermia was aban-
hypothermia is unknown. There is now evidence from
doned due to lack of effect and safety concerns (Shankaran
cohort studies that some infants with mild HIE as defined
et al., 2014, 2017). The adjusted risk ratio for death in the
using clinical criteria only in the first 6 h of life have
neonatal intensive care unit after cooling for 120 h com-
some risk of disability. The exact results have been rather
pared to 72h was 1.37 (95% confidence interval (CI):
variable, likely because of variable criteria for “mild,”
0.92, 2.04) and for 32°C compared to 33.5°C was 1.24
retrospective identification, less formal neurologic
(CI: 0.69, 2.25). Further, there was no significant overall
examinations than used in the prospective trials, or not
effect of longer or deeper cooling on death or disability
using aEEG criteria. It is critical to appreciate that some
at a mean age of 18 months (Shankaran et al., 2017).
of these infants would very likely have been classified as
These studies suggest that current protocols of cooling
having evolved to Stage II (moderate) encephalopathy by
to 33.5°C for 72 h are reasonably close to optimal. Thus
Sarnat and Sarnat by 24 h (Sarnat and Sarnat, 1976),
the most effective way to further improve outcomes from
which was based on longitudinal assessment of neuro-
therapeutic hypothermia in infants with HIE is to initiate
logic progress until hospital discharge or death plus mul-
cooling earlier, as soon as possible in the first 6 h after
timodal assessment, typically including a formal EEG
birth. Alternatively, cooling plus other pharmacologic
and imaging. Thus, at present, it is not really possible
neuroprotective agents may enable further improvements
to accurately define mild HIE in the first 6 h of life.
in outcome. There is currently considerable interest in
It is also important to appreciate that the definition of
such options, based on the endogenous induction of
mild HIE varied between studies. The CoolCap and
potentially neuroprotective compounds in the body as
TOBY studies simply specified the clinical Sarnat cri-
well as exogenous agents (Davidson et al., 2018a).
teria on a gestalt basis, but also required that infants have
moderate to severe changes on an aEEG recording before
randomization (Gluckman et al., 2005; Azzopardi et al.,
Is there benefit from cooling more than 6 h
2009). Interestingly, the NICHD trial, which used only
after birth?
clinical criteria, effectively excluded infants who
The preclinical and clinical studies reviewed previously did not have at least three criteria that are considered
consistently suggest that hypothermia should be started to be moderate/severe signs of encephalopathy
as early as possible in the first 6 h of life to achieve opti- (Shankaran et al., 2005). Thus, infants with 1 or 2 mod-
mal outcomes. However, some infants are unable to be erate or severe criteria were defined as having “mild”
started within this time window, because of late diagnosis HIE. This suggests that some of these infants actually
or being outborn in areas that cannot provide support for had “moderate” HIE even though they were excluded
cooling. Even though it is not optimal, should these from the trial, and this raises the important possibility that
infants be offered therapeutic hypothermia after 6 h of it may be possible to refine the clinical criteria to more
life? A recent RCT conducted by the Neonatal Research reliably identify infants who are at risk of disability.
Network centers in the United States compared 83 term Alternatively, potentially adding aEEG criteria might
infants who were cooled starting 6–24 h after birth at also improve the reliability of assessment.
mean  SD 16  5 h, and 85 noncooled infants (range, One meta-analysis of studies with well-defined HIE
36.5–37.3°C). Death or moderate to severe disability grading at birth and standardized neurodevelopmental
were seen in 24.4% of cooled infants, compared to assessment at 18 months or older suggested that
27.9% of noncooled infants. This difference is not signif- 86/341 (25%) of infants with “mild” HIE in the first
icant (P ¼ 0.23) (Laptook et al., 2017). The authors sug- 6 h of life had an adverse outcome (Conway et al.,
gest, using a Bayesian analysis that there was a 76% 2018). Adverse outcome was defined as death, cerebral
probability of reduced death or disability, with a small palsy, or neurodevelopmental test scores that were more
benefit of 1%–3%. Given the very small possible effect than 1 standard deviation below the mean. Although
size and that potential harm was not quantified, cooling most of these studies recruited infants solely on clinical
after 6 h cannot be recommended.It remains critical to criteria, a prospective cohort study of infants who were
focus on initiating treatment as early as possible within not treated with therapeutic hypothermia suggested that
the first 6 h after birth. infants with mild HIE, determined by both early EEG and
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 231
clinical examination, had adverse cognitive and neuro- THERAPEUTIC TARGETS FOR
motor outcomes at 5 years of age compared to healthy NEUROPROTECTION
controls (Murray et al., 2016). Although survival was
The clinical and experimental data reviewed previously
much greater after mild than moderate or severe HIE, sur-
suggest that the latent phase between the initial insult and
vivors showed no significant difference in cognitive out-
secondary energy failure corresponds with the poten-
comes between those who had had mild compared to
tially reversible activation of cell death processes. Major
moderate HIE.
initial triggers of the delayed death cascade during expo-
Given that this population of infants with “mild” HIE
sure to hypoxia–ischemia include exposure to oxygen
in the first 6 h is heterogeneous, the balance of clinical
free radical toxicity, excessive levels of excitatory amino
risk and benefit is unclear. Treating all cases of “mild”
acids, and intracellular calcium accumulation down the
HIE would lead to a considerable increase in numbers
concentration gradient due to failure of energy-
of infants being separated from their parents for at least
dependent pumps during hypoxia and opening of chan-
3 days, receiving invasive treatments such as central
nels linked to the excitatory neurotransmitters (Johnston,
lines, invasive respiratory support, sedation, and delayed
2005). However, these events rapidly resolve during
oral feeding.
reperfusion from the insult and thus cannot readily be
It is reasonable to reflect that there is evidence from
related to the effects of postinsult interventions such as
young rodents that therapeutic hypothermia seems to
cooling. It is striking that even in vitro neuronal degen-
be more protective after milder hypoxia–ischemia
eration can be prevented by cooling initiated well after
(Wassink et al., 2014). Moreover, there is evidence that
exposure to an insult (Bruno et al., 1994). Thus, the crit-
neuronal degeneration develops more slowly after a
ical effects of hypothermia must be on secondary conse-
shorter, less severe period of hypoxia–ischemia com-
quences of hypoxia–ischemia, such as the intracellular
pared with severe insults leading to infarction
progression of programmed cell death (apoptosis), the
(Beilharz et al., 1995). In adult rodents, early initiation
inflammatory reaction, and abnormal receptor activity
of cooling within 1 h and continued for 12–24 h after
(Wassink et al., 2014).
brief ischemia in adult rodents substantially reduced
CA1 necrosis at 10 and 30 days and improved learning
in an open field and T-maze after 6 months recovery
Programmed cell death
(Colbourne and Corbett, 1994, 1995). By contrast, in
adult gerbils, when the delay before initiating a 24-h There is good histologic evidence that activation of pre-
period of cooling was increased from 1 to 4 h after cere- existing programmed cell death pathways is a significant
bral ischemia, neuroprotection in the CA1 field of the contributor to posthypoxic cell death in the developing
hippocampus at 6 months of recovery fell from 70% to human brain (Thornton et al., 2017). We now know that
12% (Colbourne and Corbett, 1995). Neuroprotection posthypoxic–ischemic cell death is not purely apoptotic,
could be almost completely restored by extending the but rather includes elements of both apoptotic and
interval of moderate hypothermia to 48 h plus, even when necrotic processes, with one or the other being most
the start of cooling was delayed until 6 h after reperfusion prominent depending on factors such as maturity and
(Colbourne et al., 2000). the severity of insult (Northington et al., 2007). Consis-
Taken as a whole, these data strongly support that tent with the hypothesis that apoptotic processes are a
established protocols for therapeutic hypothermia are key therapeutic target, postinsult hypothermia started
likely to even more effectively reduce cell loss in infants after severe hypoxia–ischemia was reported to reduce
with milder clinical HIE in the first 6 h of life than those apoptotic cell death, but not necrotic cell death, in the
who were included in the trials. Speculatively, the data piglet (Edwards et al., 1995). Similarly, protection with
also suggest the intriguing hypothesis that a shorter posthypoxic–ischemic hypothermia in fetal sheep has
interval of 24 h of therapeutic hypothermia might be been closely linked with suppression of activated
beneficial for mild HIE, provided that it could be initi- caspase-3 (Bennet et al., 2007).
ated shortly after birth. Alternatively, it is possible that Encouragingly, a phase II double-blinded, placebo-
early treatment with neuroprotective agents such as controlled randomized trial of multiple dose treatment
erythropoietin that are already in clinical trial might with recombinant erythropoietin (rEpo, 1000 U/kg intra-
be effective as monotherapy for mild HIE (Wu et al., venously, at 1, 2, 3, 5, and 7 days of age) or placebo in
2016). Given that there are roughly as many infants with infants with moderate to severe HIE receiving therapeu-
mild HIE as there are with moderate to severe HIE, it is tic hypothermia suggested additional benefit (Wu et al.,
critical that the benefits of treatment for this group 2016). Infants receiving rEpo had significantly reduced
should be formally tested. brain injury scores on MRI at a mean of 5.1 days and less
232 A.J. GUNN AND M. THORESEN
frequent moderate to severe injury in multiple regions, evidence that it can activate prosurvival kinases, such
with improved motor scores at 1 year of age. Phase 3 as p-Akt and the antiapoptotic factor Bcl-2, and poten-
studies to confirm these findings are in progress. tially inhibit opening of the mitochondrial permeability
pore (Lobo et al., 2013). There is animal evidence of
Inflammatory second messengers an additive neuroprotective effect when adding xenon
to hypothermia treatment. Xenon and hypothermia
Brain injury leads to induction of the inflammatory cas-
administered together, immediately or as late as 5 h after
cade with increased release of cytokines (Hagberg et al.,
HI, in neonatal rats significantly reduced apoptotic cell
2005). These compounds are believed to exacerbate
death and loss of brain matter while improving long-term
delayed injury, whether by direct neurotoxicity and induc-
neurologic motor function and coordination (Robertson
tion of apoptosis or by promoting stimulation of leukocyte
et al., 2012). In the newborn piglet, the combination of
adhesion and infiltration into the ischemic brain. Experi-
xenon with whole body cooling was associated with a
mentally, cooling can potently suppress this inflammatory
75% reduction in global neuropathology after perinatal
reaction (Gunn and Thoresen, 2006). For example,
asphyxia (Chakkarapani et al., 2010). In a similar para-
in vitro, hypothermia inhibits proliferation, superoxide,
digm, others found that xenon-augmented hypothermia
and nitric oxide production by cultured microglia, and
reduced cerebral MRS abnormalities and cell death
in adult rats, hypothermia suppresses the posttraumatic
markers in some brain regions compared with no treat-
release of interleukin-1b and accumulation of polymor-
ment, although the effect was not significant compared
phonuclear leukocytes. Similarly, neuroprotection with
to hypothermia alone (Faulkner et al., 2011). A recent
postinsult hypothermia was associated with suppression
preliminary randomized controlled study confirmed that
of microglial activation in fetal sheep (Roelfsema et al.,
it is feasible to treat infants with xenon during therapeutic
2004; Bennet et al., 2007; Davidson et al., 2015b).
hypothermia, using up to 50% xenon for up to 18 h dur-
Other receptor and nonreceptor-mediated toxic fac-
ing cooling, with no apparent adverse effects seen at
tors are likely to contribute to neural injury in the latent
18 months follow-up (Dingley et al., 2014).
phase. For example, there is some evidence from the pre-
A subsequent study found no improvement in magnetic
term fetal sheep for delayed production of oxygen free
resonance spectroscopy and MRI biomarkers of outcome
radicals after hypoxia–ischemia (Welin et al., 2005),
measured within 15 days of birth, but was limited by rel-
which may particularly be associated with death of
atively low dose and late initiation of xenon therapy
oligodendroglia.
(Azzopardi et al., 2016).
Excitotoxity after hypoxia–ischemia
CONCLUSION
Pathologically elevated levels of extracellular excitatory
amino acids such as glutamate are seen in biphasic Therapeutic hypothermia is now established as standard
pattern. The initial increase occurs during hypoxia– care to improve neurologic recovery in infants with mod-
ischemia, which resolves rapidly after reperfusion to erate to severe HIE. However, current protocols are only
control values (Tan et al., 1996). Levels remain low partially effective (Jacobs et al., 2013). Further improve-
throughout the latent phase and then secondarily rise many ments in neurodevelopmental outcomes are likely to
hours later, in association with delayed seizures and come from combining hypothermia with endogenous
cytotoxic edema (Tan et al., 1996; Fraser et al., 2008). In or exogenous neuroprotective agents. Endogenous neu-
term-equivalent fetal sheep, for example, intense seizures roprotective compounds such as EPO and melatonin are
are seen from approximately 9  2 to 30  3 h after cerebral showing particular promise, with multiple potential ben-
ischemia; completely suppressing these large amplitude efits and excellent safety records in other settings. Tanta-
seizures with a selective glutamate antagonist was asso- lizingly, autologous or external stem cells may have
ciated with very limited reduction in neuronal damage in potential to promote long-term neurorepair through
more mildly affected regions, but no effect on infarction reducing neural inflammation and promoting release of
of the parasagittal cortex and no improvement in recovery trophic factors (Fleiss et al., 2014; van den Heuij et al.,
of EEG activity (Tan et al., 1992). Moreover, combined 2017; Bennet et al., 2018).
antiglutamate treatment and mild whole body hypother- While awaiting the results of further research, it is
mia after severe asphyxia did not show additive neuro- important not to forget that the most effective way to opti-
protection in fetal sheep (George et al., 2012). mize treatment with hypothermia is to start treatment as
The noble gas xenon provides competitive binding at soon as possible after resuscitation (Gunn and Thoresen,
the glycine binding site of the NMDA subtype of gluta- 2015). EEG recordings and other early biomarkers can
mate receptor (Dickinson et al., 2007) and so potentially help to identify patients who would benefit from treatment
can attenuate excitotoxicity. Moreover, there is some in such a limited time frame (Bennet et al., 2010).
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 233
ACKNOWLEDGMENTS Bennet L, Booth L, Gunn AJ (2010). Potential biomarkers for
hypoxic–ischemic encephalopathy. Semin Fetal Neonatal
The authors’ work reported in this review has been sup- Med 15: 253–260.
ported by grants from the Health Research Council of Bennet L, Dhillon S, Lear CA et al. (2018). Chronic inflamma-
New Zealand, Lottery Health Board of New Zealand, tion and impaired development of the preterm brain.
the Auckland Medical Research Foundation, The J Reprod Immunol 125: 45–55.
Norwegian Research Council, The Wellcome Trust, Blood AB, Hunter CJ, Power GG (2003). Adenosine mediates
Charitable donation through SPARKS UK, and the decreased cerebral metabolic rate and increased cerebral
Moulton Foundation. blood flow during acute moderate hypoxia in the near-term
fetal sheep. J Physiol 553: 935–945.
Blumberg RM, Cady EB, Wigglesworth JS et al. (1997).
REFERENCES
Relation between delayed impairment of cerebral energy
Alonso-Alconada D, Broad KD, Bainbridge A et al. (2015). metabolism and infarction following transient focal hyp-
Brain cell death is reduced with cooling by 3.5 degrees oxia–ischaemia in the developing brain. Exp Brain Res
C to 5 degrees C but increased with cooling by 8.5 degrees 113: 130–137.
C in a piglet asphyxia model. Stroke 46: 275–278. Boddy K, Dawes GS, Fisher R et al. (1974). Foetal respiratory
Azzopardi D, Wyatt JS, Cady EB et al. (1989). Prognosis of movements, electrocortical and cardiovascular responses
newborn infants with hypoxic–ischemic brain injury to hypoxaemia and hypercapnia in sheep. J Physiol 243:
assessed by phosphorus magnetic resonance spectroscopy. 599–618.
Pediatr Res 25: 445–451. Brar HS, Platt LD, DeVore GR et al. (1988). Qualitative
Azzopardi DV, Strohm B, Edwards AD et al. (2009). Moderate assessment of maternal uterine and fetal umbilical
hypothermia to treat perinatal asphyxial encephalopathy. artery blood flow and resistance in laboring patients
N Engl J Med 361: 1349–1358. by Doppler velocimetry. Am J Obstet Gynecol 158:
Azzopardi D, Strohm B, Marlow N et al. (2014). Effects of 952–956.
hypothermia for perinatal asphyxia on childhood out- Bruno VM, Goldberg MP, Dugan LL et al. (1994).
comes. N Engl J Med 371: 140–149. Neuroprotective effect of hypothermia in cortical cultures
Azzopardi D, Robertson NJ, Bainbridge A et al. (2016). exposed to oxygen-glucose deprivation or excitatory amino
Moderate hypothermia within 6 h of birth plus inhaled acids. J Neurochem 63: 1398–1406.
xenon versus moderate hypothermia alone after birth Chakkarapani E, Dingley J, Liu X et al. (2010). Xenon
asphyxia (TOBY-Xe): a proof-of-concept, open-label, ran- enhances hypothermic neuroprotection in asphyxiated
domised controlled trial. Lancet Neurol 15: 145–153. newborn pigs. Ann Neurol 68: 330–341.
Basu SK, Kaiser JR, Guffey D et al. (2016). Hypoglycaemia Colbourne F, Corbett D (1994). Delayed and prolonged post-
and hyperglycaemia are associated with unfavourable out- ischemic hypothermia is neuroprotective in the gerbil.
come in infants with hypoxic ischaemic encephalopathy: a Brain Res 654: 265–272.
post hoc analysis of the CoolCap Study. Arch Dis Child Colbourne F, Corbett D (1995). Delayed postischemic hypo-
Fetal Neonatal Ed 101: F149–F155. thermia: a six month survival study using behavioral and
Bax M, Nelson KB (1993). Birth asphyxia: a statement. World histological assessments of neuroprotection. J Neurosci
Federation of Neurology Group. Dev Med Child Neurol 35: 15: 7250–7260.
1022–1024. Colbourne F, Auer RN, Sutherland GR (1998).
Beilharz EJ, Williams CE, Dragunow M et al. (1995). Characterization of postischemic behavioral deficits in ger-
Mechanisms of delayed cell death following hypoxic– bils with and without hypothermic neuroprotection. Brain
ischemic injury in the immature rat: evidence for apoptosis Res 803: 69–78.
during selective neuronal loss. Brain Res Mol Brain Res 29: Colbourne F, Corbett D, Zhao Z et al. (2000). Prolonged but
1–14. delayed postischemic hypothermia: a long-term outcome
Bennet L (2017). Sex, drugs and rock and roll: tales from pre- study in the rat middle cerebral artery occlusion model.
term fetal life. J Physiol 595: 1865–1881. J Cereb Blood Flow Metab 20: 1702–1708.
Bennet L, Peebles DM, Edwards AD et al. (1998). The cerebral Conway JM, Walsh BH, Boylan GB et al. (2018). Mild hyp-
hemodynamic response to asphyxia and hypoxia in the oxic ischaemic encephalopathy and long term neurodeve-
near-term fetal sheep as measured by near infrared spec- lopmental outcome—a systematic review. Early Hum
troscopy. Pediatr Res 44: 951–957. Dev 120: 80–87.
Bennet L, Roelfsema V, Pathipati P et al. (2006). Relationship Cowan F, Rutherford M, Groenendaal F et al. (2003). Origin
between evolving epileptiform activity and delayed loss of and timing of brain lesions in term infants with neonatal
mitochondrial activity after asphyxia measured by near- encephalopathy. Lancet 361: 736–742.
infrared spectroscopy in preterm fetal sheep. J Physiol Davidson JO, Green CR, Bennet L et al. (2015a). Battle of the
572: 141–154. hemichannels—connexins and pannexins in ischemic brain
Bennet L, Roelfsema V, George S et al. (2007). The effect of injury. Int J Dev Neurosci 45: 66–74.
cerebral hypothermia on white and grey matter injury Davidson JO, Wassink G, Yuill CA et al. (2015b). How long is
induced by severe hypoxia in preterm fetal sheep. too long for cerebral cooling after ischemia in fetal sheep?
J Physiol 578: 491–506. J Cereb Blood Flow Metab 35: 751–758.
234 A.J. GUNN AND M. THORESEN
Davidson JO, Dean JM, Fraser M et al. (2018a). Perinatal brain George SA, Barrett RD, Bennet L et al. (2012). Nonadditive
injury: mechanisms and therapeutic approaches. Front neuroprotection with early glutamate receptor blockade
Biosci (Landmark Ed) 23: 2204–2226. and delayed hypothermia after asphyxia in preterm fetal
Davidson JO, Draghi V, Whitham S et al. (2018b). How long is sheep. Stroke 43: 3114–3117.
sufficient for optimal neuroprotection with cerebral cool- Gilbert RD, Schroder H, Kawamura T et al. (1985). Heat trans-
ing after ischemia in fetal sheep? J Cereb Blood Flow fer pathways between fetal lamb and ewe. J Appl Physiol
Metab 38: 1047–1059. 59: 634–638.
de Haan HH, van Reempts JL, Vles JS et al. (1993). Effects of Giussani DA, Spencer JA, Moore PJ et al. (1993). Afferent
asphyxia on the fetal lamb brain. Am J Obstet Gynecol 169: and efferent components of the cardiovascular reflex
1493–1501. responses to acute hypoxia in term fetal sheep. J Physiol
de Haan HH, Gunn AJ, Williams CE et al. (1997a). Brief 461: 431–449.
repeated umbilical cord occlusions cause sustained cyto- Glass HC, Shellhaas RA, Wusthoff CJ et al. (2016).
toxic cerebral edema and focal infarcts in near-term fetal Contemporary profile of seizures in neonates: a prospective
lambs. Pediatr Res 41: 96–104. cohort study. J Pediatr 174: 98–103 e101.
de Haan HH, Gunn AJ, Williams CE et al. (1997b). Glass HC, Shellhaas RA, Tsuchida TN et al. (2017). Seizures
Magnesium sulfate therapy during asphyxia in near-term in preterm neonates: a multicenter observational cohort
fetal lambs does not compromise the fetus but does not study. Pediatr Neurol 72: 19–24.
reduce cerebral injury. Am J Obstet Gynecol 176: 18–27. Gluckman PD, Wyatt JS, Azzopardi D et al. (2005). Selective
Deng W, Yue Q, Rosenberg PA et al. (2006). Oligodendrocyte head cooling with mild systemic hypothermia after neona-
excitotoxicity determined by local glutamate accumulation tal encephalopathy: multicentre randomised trial. Lancet
and mitochondrial function. J Neurochem 98: 213–222. 365: 663–670.
Dickinson R, Peterson BK, Banks P et al. (2007). Competitive Green LR, Bennet L, Hanson MA (1996). The role of nitric
inhibition at the glycine site of the N-methyl-D-aspartate oxide synthesis in cardiovascular responses to acute hypoxia
receptor by the anesthetics xenon and isoflurane: evidence in the late gestation sheep fetus. J Physiol 497: 271–277.
from molecular modeling and electrophysiology. Greenwell EA, Wyshak G, Ringer SA et al. (2012).
Anesthesiology 107: 756–767. Intrapartum temperature elevation, epidural use, and
Dingley J, Tooley J, Liu X et al. (2014). Xenon ventilation dur- adverse outcome in term infants. Pediatrics 129:
ing therapeutic hypothermia in neonatal encephalopathy: a e447–e454.
feasibility study. Pediatrics 133: 809–818. Guiang 3rd SF, Widness JA, Flanagan KB et al. (1993). The
Edwards AD, Yue X, Squier MV et al. (1995). Specific inhi- relationship between fetal arterial oxygen saturation and
bition of apoptosis after cerebral hypoxia–ischaemia by heart and skeletal muscle myoglobin concentrations in
moderate post-insult hypothermia. Biochem Biophys Res the ovine fetus. J Dev Physiol 19: 99–104.
Commun 217: 1193–1199. Guillet R, Edwards AD, Thoresen M et al. (2012). Seven- to
Eklind S, Mallard C, Arvidsson P et al. (2005). eight-year follow-up of the CoolCap trial of head
Lipopolysaccharide induces both a primary and a second- cooling for neonatal encephalopathy. Pediatr Res 71:
ary phase of sensitization in the developing rat brain. 205–209.
Pediatr Res 58: 112–116. Gunn AJ, Bennet L (2009). Fetal hypoxia insults and patterns
Faulkner S, Bainbridge A, Kato T et al. (2011). Xenon aug- of brain injury: insights from animal models. Clin Perinatol
mented hypothermia reduces early lactate/N- 36: 579–593.
acetylaspartate and cell death in perinatal asphyxia. Ann Gunn AJ, Thoresen M (2006). Hypothermic neuroprotection.
Neurol 70: 133–150. NeuroRx 3: 154–169.
Fisher DJ, Heymann MA, Rudolph AM (1982). Fetal myocar- Gunn AJ, Thoresen M (2015). Animal studies of neonatal
dial oxygen and carbohydrate consumption during acutely hypothermic neuroprotection have translated well in to
induced hypoxemia. Am J Physiol 242: H657–H661. practice. Resuscitation 97: 88–90.
Fleischer A, Anyaegbunam AA, Schulman H et al. (1987). Gunn AJ, Parer JT, Mallard EC et al. (1992). Cerebral histo-
Uterine and umbilical artery velocimetry during normal logic and electrocorticographic changes after asphyxia in
labor. Am J Obstet Gynecol 157: 40–43. fetal sheep. Pediatr Res 31: 486–491.
Fleiss B, Gressens P (2012). Tertiary mechanisms of brain Gunn AJ, Gunn TR, de Haan HH et al. (1997). Dramatic neu-
damage: a new hope for treatment of cerebral palsy? ronal rescue with prolonged selective head cooling after
Lancet Neurol 11: 556–566. ischemia in fetal lambs. J Clin Invest 99: 248–256.
Fleiss B, Guillot PV, Titomanlio L et al. (2014). Stem cell ther- Gunn AJ, Gunn TR, Gunning MI et al. (1998). Neuroprotection
apy for neonatal brain injury. Clin Perinatol 41: 133–148. with prolonged head cooling started before postischemic
Fraser M, Bennet L, van Zijl PL et al. (2008). Extracellular seizures in fetal sheep. Pediatrics 102: 1098–1106.
amino acids and peroxidation products in the periventricu- Gunn AJ, Bennet L, Gunning MI et al. (1999). Cerebral hypo-
lar white matter during and after cerebral ischemia in pre- thermia is not neuroprotective when started after postis-
term fetal sheep. J Neurochem 105: 2214–2223. chemic seizures in fetal sheep. Pediatr Res 46: 274–280.
Galinsky R, Lear CA, Dean JM et al. (2018). Complex inter- Hagberg H, Mallard C, Jacobsson B (2005). Role of cytokines
actions between hypoxia–ischemia and inflammation in in preterm labour and brain injury. BJOG 112 (Suppl. 1):
preterm brain injury. Dev Med Child Neurol 60: 126–133. 16–18.
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 235
Hagberg H, Mallard C, Ferriero DM et al. (2015). The role of Li J, Takeda Y, Hirakawa M (2000). Threshold of ischemic
inflammation in perinatal brain injury. Nat Rev Neurol 11: depolarization for neuronal injury following four-vessel
192–208. occlusion in the rat cortex. J Neurosurg Anesthesiol 12:
Harris DL, Weston PJ, Harding JE (2015). Lactate, rather than 247–254.
ketones, may provide alternative cerebral fuel in hypogly- Li H, Gudmundsson S, Olofsson P (2003). Uterine artery blood
caemic newborns. Arch Dis Child Fetal Neonatal Ed 100: flow velocity waveforms during uterine contractions.
F161–F164. Ultrasound Obstet Gynecol 22: 578–585.
Huch A, Huch R, Schneider H et al. (1977). Continuous trans- Liu X, Jary S, Cowan F et al. (2017). Reduced infancy and
cutaneous monitoring of fetal oxygen tension during childhood epilepsy following hypothermia-treated neona-
labour. Br J Obstet Gynaecol 84: 1–39. tal encephalopathy. Epilepsia 58: 1902–1911.
Hunter CJ, Bennet L, Power GG et al. (2003). Key neuroprotec- Lobo N, Yang B, Rizvi M et al. (2013). Hypothermia and
tive role for endogenous adenosine A1 receptor activation xenon: novel noble guardians in hypoxic–ischemic enceph-
during asphyxia in the fetal sheep. Stroke 34: 2240–2245. alopathy? J Neurosci Res 91: 473–478.
Ikeda T, Murata Y, Quilligan EJ et al. (1998). Physiologic and Longo LD, Koos BJ, Power GG (1973). Fetal myoglobin:
histologic changes in near-term fetal lambs exposed to quantitative determination and importance for oxygena-
asphyxia by partial umbilical cord occlusion. Am tion. Am J Physiol 224: 1032–1036.
J Obstet Gynecol 178: 24–32. Lorek A, Takei Y, Cady EB et al. (1994). Delayed ("second-
Impey LW, Greenwood CE, Black RS et al. (2008). The rela- ary") cerebral energy failure after acute hypoxia–ischemia
tionship between intrapartum maternal fever and neonatal in the newborn piglet: continuous 48-hour studies by phos-
acidosis as risk factors for neonatal encephalopathy. Am phorus magnetic resonance spectroscopy. Pediatr Res 36:
J Obstet Gynecol 198: 49 e41–46. 699–706.
Jacobs SE, Berg M, Hunt R et al. (2013). Cooling for newborns Maitre NL (2015). Neurorehabilitation after neonatal intensive
with hypoxic ischaemic encephalopathy. Cochrane care: evidence and challenges. Arch Dis Child Fetal
Database Syst Rev 1: CD003311. Neonatal Ed 100: F534–F540.
Janbu T, Nesheim BI (1987). Uterine artery blood velocities Mallard EC, Gunn AJ, Williams CE et al. (1992). Transient
during contractions in pregnancy and labour related to umbilical cord occlusion causes hippocampal damage in
intrauterine pressure. Br J Obstet Gynaecol 94: 1150–1155. the fetal sheep. Am J Obstet Gynecol 167: 1423–1430.
Jary S, Smit E, Liu X et al. (2015). Less severe cerebral palsy Mallard EC, Williams CE, Johnston BM et al. (1995).
outcomes in infants treated with therapeutic hypothermia. Repeated episodes of umbilical cord occlusion in fetal
Acta Paediatr 104: 1241–1247. sheep lead to preferential damage to the striatum and sen-
Johnston MV (2005). Excitotoxicity in perinatal brain injury. sitize the heart to further insults. Pediatr Res 37: 707–713.
Brain Pathol 15: 234–240. Martinez-Biarge M, Cheong JL, Diez-Sebastian J et al. (2016).
Jonsson M, Agren J, Norden-Lindeberg S et al. (2014). Risk factors for neonatal arterial ischemic stroke: the impor-
Neonatal encephalopathy and the association to asphyxia tance of the intrapartum period. J Pediatr 173: 62–68 e61.
in labor. Am J Obstet Gynecol 211: 667 e661–668. McIntosh GH, Baghurst KI, Potter BJ et al. (1979). Foetal
Katz M, Lunenfeld E, Meizner I et al. (1987). The effect of the brain development in the sheep. Neuropathol Appl
duration of the second stage of labour on the acid-base state Neurobiol 5: 103–114.
of the fetus. Br J Obstet Gynaecol 94: 425–430. Miller SP, Ramaswamy V, Michelson D et al. (2005). Patterns
Keogh MJ, Drury PP, Bennet L et al. (2012). Limited predic- of brain injury in term neonatal encephalopathy. J Pediatr
tive value of early changes in EEG spectral power for neu- 146: 453–460.
ral injury after asphyxia in preterm fetal sheep. Pediatr Res Modanlou H, Yeh SY, Hon EH (1974). Fetal and neonatal
71: 345–353. acid–base balance in normal and high-risk pregnancies:
Laptook AR, Shankaran S, Tyson JE et al. (2017). Effect of during labor and the first hour of life. Obstet Gynecol 43:
therapeutic hypothermia initiated after 6 hours of age on 347–353.
death or disability among newborns with hypoxic-ischemic Morishima HO, Glaser B, Niemann WH et al. (1975). Increased
encephalopathy: A randomized clinical trial. JAMA 318: uterine activity and fetal deterioration during maternal
1550–1560. hyperthermia. Am J Obstet Gynecol 121: 531–538.
Lear CA, Wassink G, Westgate JA et al. (2018). The Murray DM, O’Connor CM, Ryan CA et al. (2016). Early EEG
peripheral chemoreflex—indefatigable guardian of grade and outcome at 5 years after mild neonatal hypoxic
fetal physiological adaptation to labour. J Physiol 596: ischemic encephalopathy. Pediatrics 138: e20160659.
5611–5623. (ePub).
Lee AC, Kozuki N, Blencowe H et al. (2013). Intrapartum- Niquet J, Seo DW, Allen SG et al. (2006). Hypoxia in presence
related neonatal encephalopathy incidence and impairment of blockers of excitotoxicity induces a caspase-dependent
at regional and global levels for 2010 with trends from neuronal necrosis. Neuroscience 141: 77–86.
1990. Pediatr Res 74: 50–72. Northington FJ, Zelaya ME, O’Riordan DP et al. (2007). Failure
Lemyre B, Ly L, Chau V et al. (2017). Initiation of passive to complete apoptosis following neonatal hypoxia–ischemia
cooling at referring centre is most predictive of achieving manifests as "continuum" phenotype of cell death and occurs
early therapeutic hypothermia in asphyxiated newborns. with multiple manifestations of mitochondrial dysfunction
Paediatr Child Health 22: 264–268. in rodent forebrain. Neuroscience 149: 822–833.
236 A.J. GUNN AND M. THORESEN
Novak I, Morgan C, Adde L et al. (2017). Early, accurate diag- Shankaran S, Laptook AR, Ehrenkranz RA et al. (2005).
nosis and early intervention in cerebral palsy: advances in Whole-body hypothermia for neonates with hypoxic–
diagnosis and treatment. JAMA Pediatr 171: 897–907. ischemic encephalopathy. N Engl J Med 353: 1574–1584.
Odd DE, Gunnell D, Lewis G et al. (2011). Long-term impact Shankaran S, Pappas A, McDonald SA et al. (2012). Childhood
of poor birth condition on social and economic outcomes in outcomes after hypothermia for neonatal encephalopathy.
early adulthood. Pediatrics 127: e1498–e1504. N Engl J Med 366: 2085–2092.
Osredkar D, Thoresen M, Maes E et al. (2014). Hypothermia Shankaran S, Laptook AR, Pappas A et al. (2014). Effect of
is not neuroprotective after infection-sensitized neonatal depth and duration of cooling on deaths in the NICU among
hypoxic–ischemic brain injury. Resuscitation 85: 567–572. neonates with hypoxic ischemic encephalopathy: a ran-
Parer JT (1998). Effects of fetal asphyxia on brain cell struc- domized clinical trial. JAMA 312: 2629–2639.
ture and function: limits of tolerance. Comp Biochem Shankaran S, Laptook AR, Pappas A et al. (2017). Effect of
Physiol A Mol Integr Physiol 119: 711–716. depth and duration of cooling on death or disability at age
Peebles DM, Spencer JA, Edwards AD et al. (1994). Relation 18 months among neonates with hypoxic–ischemic enceph-
between frequency of uterine contractions and human fetal alopathy: a randomized clinical trial. JAMA 318: 57–67.
cerebral oxygen saturation studied during labour by near Sinding M, Peters DA, Frokjaer JB et al. (2016). Reduced pla-
infrared spectroscopy. Br J Obstet Gynaecol 101: 44–48. cental oxygenation during subclinical uterine contractions
Peeters LL, Sheldon RE, Jones Jr MD et al. (1979). Blood flow as assessed by BOLD MRI. Placenta 39: 16–20.
to fetal organs as a function of arterial oxygen content. Am Sirimanne ES, Blumberg RM, Bossano D et al. (1996). The
J Obstet Gynecol 135: 637–646. effect of prolonged modification of cerebral temperature
Pitcher JB, Robertson AL, Cockington RA et al. (2009). on outcome after hypoxic–ischemic brain injury in the
Prenatal growth and early postnatal influences on adult infant rat. Pediatr Res 39: 591–597.
motor cortical excitability. Pediatrics 124: e128–e136. Spain JE, Tuuli MG, Macones GA et al. (2015). Risk factors
Ramsey EM (1968). Uteroplacental circulation during labor. for serious morbidity in term nonanomalous neonates.
Clin Obstet Gynaecol 11: 78–95. Am J Obstet Gynecol 212: 799 e791–797.
Rees S, Mallard C, Breen S et al. (1998). Fetal brain injury fol- Tan WK, Williams CE, Gunn AJ et al. (1992). Suppression of
lowing prolonged hypoxemia and placental insufficiency: a postischemic epileptiform activity with MK-801 improves
review. Comp Biochem Physiol A Mol Integr Physiol 119: neural outcome in fetal sheep. Ann Neurol 32: 677–682.
653–660. Tan WK, Williams CE, During MJ et al. (1996). Accumulation
Reid SM, Meehan E, McIntyre S et al. (2016). Temporal trends of cytotoxins during the development of seizures and
in cerebral palsy by impairment severity and birth gesta- edema after hypoxic–ischemic injury in late gestation fetal
tion. Dev Med Child Neurol 58 (Suppl. 2): 25–35. sheep. Pediatr Res 39: 791–797.
Reuss ML, Rudolph AM, Heymann MA (1981). Selective dis- Thoresen M, Penrice J, Lorek A et al. (1995). Mild hypother-
tribution of microspheres injected into the umbilical veins mia after severe transient hypoxia–ischemia ameliorates
and inferior venae cavae of fetal sheep. Am J Obstet delayed cerebral energy failure in the newborn piglet.
Gynecol 141: 427–432. Pediatr Res 37: 667–670.
Robertson CM, Finer NN, Grace MG (1989). School perfor- Thoresen M, Tooley J, Liu X et al. (2013). Time is brain: start-
mance of survivors of neonatal encephalopathy associated ing therapeutic hypothermia within three hours after birth
with birth asphyxia at term. J Pediatr 114: 753–760. improves motor outcome in asphyxiated newborns.
Robertson NJ, Tan S, Groenendaal F et al. (2012). Which neu- Neonatology 104: 228–233.
roprotective agents are ready for bench to bedside transla- Thornton JS, Ordidge RJ, Penrice J et al. (1998). Temporal and
tion in the newborn infant? J Pediatr 160: 544–552.e544. anatomical variations of brain water apparent diffusion
Roelfsema V, Bennet L, George S et al. (2004). The window of coefficient in perinatal cerebral hypoxic–ischemic injury:
opportunity for cerebral hypothermia and white matter relationships to cerebral energy metabolism. Magn Reson
injury after cerebral ischemia in near-term fetal sheep. Med 39: 920–927.
J Cereb Blood Flow Metab 24: 877–886. Thornton C, Leaw B, Mallard C et al. (2017). Cell death in the
Roth SC, Baudin J, Cady E et al. (1997). Relation of deranged developing brain after hypoxia–ischemia. Front Cell
neonatal cerebral oxidative metabolism with neurodeve- Neurosci 11: 248.
lopmental outcome and head circumference at 4 years. Tooley JR, Satas S, Porter H et al. (2003). Head cooling with
Dev Med Child Neurol 39: 718–725. mild systemic hypothermia in anesthetized piglets is neuro-
Sabir H, Scull-Brown E, Liu X et al. (2012). Immediate hypo- protective. Ann Neurol 53: 65–72.
thermia is not neuroprotective after severe hypoxia– Torvik A (1984). The pathogenesis of watershed infarcts in the
ischemia and is deleterious when delayed by 12 hours in brain. Stroke 15: 221–223.
neonatal rats. Stroke 43: 3364–3370. Tsuji M, Naruse H, Volpe J et al. (1995). Reduction of cyto-
Sarnat HB, Sarnat MS (1976). Neonatal encephalopathy fol- chrome aa3 measured by near-infrared spectroscopy predicts
lowing fetal distress. A clinical and electroencephalo- cerebral energy loss in hypoxic piglets. Pediatr Res 37:
graphic study. Arch Neurol 33: 696–705. 253–259.
Sato M, Noguchi J, Mashima M et al. (2016). 3D power Turbow RM, Curran-Everett D, Hay Jr WW et al. (1995).
Doppler ultrasound assessment of placental perfusion dur- Cerebral lactate metabolism in near-term fetal sheep. Am
ing uterine contraction in labor. Placenta 45: 32–36. J Physiol 269: R938–R942.
NEONATAL ENCEPHALOPATHY AND HYPOXIC–ISCHEMIC ENCEPHALOPATHY 237
van den Heuij LG, Fraser M, Miller SL et al. (2017). Delayed repeated cord occlusion in near-term fetal sheep. Am
intranasal infusion of human amnion epithelial cells J Obstet Gynecol 193: 1526–1533.
improves white matter maturation after asphyxia in preterm Westgate JA, Wibbens B, Bennet L et al. (2007). The intrapar-
fetal sheep. J Cereb Blood Flow Metab 39 (2): 223–239. tum deceleration in center stage: a physiological approach
Epub Sept: 271678X17729954. to interpretation of fetal heart rate changes in labor. Am
Vannucci RC (2000). Hypoxic–ischemic encephalopathy. Am J Obstet Gynecol 197: 236.e1–11.
J Perinatol 17: 113–120. Wiberg N, Kallen K, Olofsson P (2006). Physiological devel-
Vannucci RC, Towfighi J, Vannucci SJ (2004). Secondary opment of a mixed metabolic and respiratory umbilical
energy failure after cerebral hypoxia–ischemia in the cord blood acidemia with advancing gestational age.
immature rat. J Cereb Blood Flow Metab 24: 1090–1097. Early Hum Dev 82: 583–589.
Vesoulis ZA, Liao SM, Rao R et al. (2017). Re-examining the Wilkening RB, Meschia G (1983). Fetal oxygen uptake, oxy-
arterial cord blood gas pH screening criteria in neonatal genation, and acid–base balance as a function of uterine
encephalopathy. Arch Dis Child Fetal Neonatal Ed 103 blood flow. Am J Physiol 244: H749–H755.
(4): F377–F382. Epub Sep 23. Williams CE, Gunn AJ, Synek B et al. (1990). Delayed sei-
Wassink G, Bennet L, Davidson JO et al. (2013). Pre-existing zures occurring with hypoxic–ischemic encephalopathy
hypoxia is associated with greater EEG suppression and in the fetal sheep. Pediatr Res 27: 561–565.
early onset of evolving seizure activity during brief repeated Williams CE, Gunn A, Gluckman PD (1991). Time course
asphyxia in near-term fetal sheep. PLoS One 8: e73895. of intracellular edema and epileptiform activity fol-
Wassink G, Gunn ER, Drury PP et al. (2014). The mechanisms lowing prenatal cerebral ischemia in sheep. Stroke 22:
and treatment of asphyxial encephalopathy. Front Neurosci 516–521.
8: 40. Winkler M, Rath W (1999). A risk-benefit assessment of oxy-
Wassink G, Davidson JO, Lear CA et al. (2018). A working tocics in obstetric practice. Drug Saf 20: 323–345.
model for hypothermic neuroprotection. J Physiol 596 Wood T, Osredkar D, Puchades M et al. (2016).
(23): 5641–5654. Treatment temperature and insult severity influence the
Welin AK, Sandberg M, Lindblom A et al. (2005). White mat- neuroprotective effects of therapeutic hypothermia. Sci
ter injury following prolonged free radical formation in the Rep 6: 23430.
0.65 gestation fetal sheep brain. Pediatr Res 58: 100–105. Wu YW, Mathur AM, Chang T et al. (2016). High-dose eryth-
Westgate JA, Gunn AJ, Gunn TR (1999). Antecedents of neo- ropoietin and hypothermia for hypoxic–ischemic encepha-
natal encephalopathy with fetal acidaemia at term. BJOG lopathy: a phase II trial. Pediatrics 137: e20160191.
106: 774–782. Wyatt JS, Edwards AD, Azzopardi D et al. (1989). Magnetic
Westgate J, Wassink G, Bennet L et al. (2005). Spontaneous resonance and near infrared spectroscopy for investigation
hypoxia in multiple pregnancy is associated with early fetal of perinatal hypoxic–ischaemic brain injury. Arch Dis
decompensation and greater T wave elevation during brief Child 64: 953–963.

You might also like