You are on page 1of 7

Clinical Review & Education

Review

Hypoxic-Ischemic Encephalopathy
A Review for the Clinician
Martha Douglas-Escobar, MD; Michael D. Weiss, MD

Supplemental content at
IMPORTANCE Hypoxic-ischemic encephalopathy (HIE) occurs in 1 to 8 per 1000 live births in jamapediatrics.com
developed countries. Historically, the clinician has had little to offer neonates with HIE other
than systemic supportive care. Recently, the neuroprotective therapy of hypothermia has
emerged as the standard of care, and other complementary therapies are rapidly
transitioning from the basic science to clinical care.

OBJECTIVE To examine the pathophysiology of HIE and the state of the art for the clinical care
of neonates with HIE.

EVIDENCE REVIEW We performed a literature review using the PubMed database. Results
focused on reviews and articles published from January 1, 2004, through December 31, 2014.
Articles published earlier than 2004 were included when appropriate for historical
perspective. Our review emphasized evidence-based management practices for the clinician.

FINDINGS A total of 102 articles for critical review were selected based on their relevance to
the incidence of HIE, pathophysiology, neuroimaging, placental pathology, biomarkers,
current systemic supportive care, hypothermia, and emerging therapies for HIE and were
reviewed by both of us. Seventy-five publications were selected for inclusion in this article
based on their relevance to these topics. The publications highlight the emergence of
serum-based biomarkers, placental pathology, and magnetic resonance imaging as useful
tools to predict long-term outcomes. Hypothermia and systemic supportive care form the
cornerstone of therapy for HIE.
Author Affiliations: Department of
Pediatrics, University of California,
CONCLUSIONS AND RELEVANCE The pathophysiology of HIE is now better understood, and San Francisco (Douglas-Escobar);
treatment with hypothermia has become the foundation of therapy. Several neuroprotective Department of Pediatrics, University
of Florida, Gainesville (Weiss).
agents offer promise when combined with hypothermia and are entering clinical trials.
Corresponding Author: Michael D.
Weiss, MD, Department of Pediatrics,
JAMA Pediatr. 2015;169(4):397-403. doi:10.1001/jamapediatrics.2014.3269 University of Florida, 1600 SW Archer
Published online February 16, 2015. Rd, PO Box 100296, Gainesville, FL
32610 (mweiss@ufl.edu).

H
ypoxic-ischemic encephalopathy (HIE) is a major cause of
neurologic disabilities in term neonates despite the re- Pathophysiology
cent widespread use of hypothermia therapy. The inci-
dence of HIE ranges from 1 to 8 per 1000 live births in developed Clinicians must understand the pathophysiology of injury during hy-
countries and is as high as 26 per 1000 live births in underdevel- poxia-ischemia (HI) to manage this critical illness in neonates ap-
oped countries.1 Although the advent of therapeutic hypothermia propriately because the injury evolves over the course of days and
offers neuroprotection, the improvement in outcomes has been possibly weeks (Figure 1).3 Furthermore, a bedside clinician who un-
modest. Therefore, new synergistic therapies are needed to im- derstands the pathophysiology of HIE will understand the mecha-
prove outcomes. This review is intended for the clinician and briefly nism of action of the various emerging neuroprotective agents.
examines the pathophysiology of HIE in the context of clinical care Adequate cerebral blood flow delivers oxygen and glucose to the
(more extensive reviews on this topic are found in Johnston et al2). fetal brain. This blood flow helps the fetal brain maintain homeosta-
This review examines practical clinical information, such as diagnos- sis and meet cellular energy demands. A variety of conditions de-
tic considerations, and emphasizes evidence-based practices for neo- crease placental perfusion or disrupt the delivery of oxygen and glu-
nates with HIE (eTable 1 in the Supplement summarizes pertinent cose in the umbilical cord, including placental abruption, prolapse of
publications from the past 5 years in each of these categories). The the umbilical cord, and uterine rupture. The hypoxia eventually leads
review examines 75 articles (of the 102 selected for critical review), to a decrease in fetal cardiac output, which reduces cerebral blood
with an emphasis on articles published between January 1, 2004, flow. If the decrease in cerebral blood flow is moderate, the cerebral
and December 21, 2014. arteries shunt blood flow from the anterior circulation to the poste-

jamapediatrics.com (Reprinted) JAMA Pediatrics April 2015 Volume 169, Number 4 397

Copyright 2015 American Medical Association. All rights reserved.

Downloaded From: http://archpedi.jamanetwork.com/ by a University of Iowa User on 05/23/2015


Clinical Review & Education Review Hypoxic-Ischemic Encephalopathy

Figure 1. Schematic Overview of the Pathophysiological Features of Hypoxic-Ischemic Encephalopathy

Latent Secondary Tertiary


Insult
Cell death
Multiple programmed Late cell death
Secondary
cell death pathways mitochondrial failure
Reperfusion
Response

Seizures Remodeling

Excitotoxicity Astrogliosis
Glucose O2 delivery
Inflammation
Repair
ATP production
Oxidative stress

30 min 6-12 h ≥3 d Months ATP indicates adenosine


triphosphate; O2, oxygen.

rior circulation to maintain adequate perfusion of the brainstem, cer- rologic dysfunction in the form of neonatal encephalopathy. Hall-
ebellum, and basal ganglia.4 As a result, damage is restricted to the marks of neonatal encephalopathy are depression of the level of con-
cerebral cortex and watershed areas of the cerebral hemispheres. sciousness, often with respiratory depression, abnormality of muscle
Acute hypoxia causes an abrupt decrease in cerebral blood flow, which tone and power, disturbances of cranial nerve function, and
produces injury to the basal ganglia and thalami.4 seizures.9 Evidence of low Apgar scores and metabolic acidosis (in
Decreased cerebral perfusion sets in motion a temporal se- arterial cord oxygen or newborn blood oxygen levels) must accom-
quence of injury, which clinicians have divided into distinct phases. pany the neurologic dysfunction.9 Metabolic acidosis strongly sug-
In the acute phase, the decreased cerebral blood flow reduces the gests HI injury. Concomitant injury to other organs, such as the liver
delivery of oxygen and glucose to the brain, which leads to anaero- (elevated transaminase level), the kidneys (elevated creatinine level),
bic metabolism. As a result, production of adenosine triphosphate and/or the heart (elevated creatine kinase–MB fraction and tro-
decreases and that of lactic acid increases. The depletion in aden- ponin T levels), provides further evidence of HI injury.10 In addi-
osine triphosphate reduces transcellular transport and leads to in- tion, the pattern of injury on magnetic resonance imaging (MRI) of
tracellular accumulation of sodium, water, and calcium.5 When the the brain may further confirm HIE.
membrane depolarizes, the cell releases the excitatory amino acid Neonates with suspected HIE are classified according to the Sar-
glutamate, and calcium flows into the cell via N-methyl-D-aspartate– nat staging system,11 which evaluates the level of consciousness,
gated channels. This cascade of events perpetuates injury in a pro- muscle tone, tendon reflexes, complex reflexes, and autonomic func-
cess termed excitotoxicity. The peroxidation of free fatty acids by oxy- tion. The Sarnat stage classifies neonatal HIE into the following 3 cat-
gen free radicals leads to more cellular damage.3 The culmination egories: stage I (mild), stage II (moderate), and stage III (severe). En-
of energy failure, acidosis, glutamate release, lipid peroxidation, and try criteria for therapeutic hypothermia include a modified version
the toxic effects of nitric oxide leads to cell death via necrosis and of the Sarnat staging system.
activates apoptotic cascades.3
Depending on the timing of injury and the degree of medical inter- Biomarkers
vention,apartialrecoveryoccursduringthe30to60minutesafterthe In neonates with HIE, monitoring and evaluation, outcome predic-
acute insult or the primary phase of injury. This partial recovery ushers tion, and response to the hypothermia treatment are measured with
in a latent phase of injury.6 The latent phase may last from 1 to 6 hours a combination of a neurologic examination, MRI, and electroen-
andischaracterizedbyrecoveryofoxidativemetabolism,inflammation, cephalography (EEG).12 However, unstable neonates may not tol-
andcontinuationoftheactivatedapoptoticcascades.5 Asecondaryde- erate transport for an MRI of the brain or the length of the MRI scan-
terioration follows the latent phase in neonates with moderate to se- ning time. Moreover, hypothermia therapy may depress the
vere injury. The secondary phase of injury occurs within approximately amplitude-integrated EEG (aEEG) and thus limit the early predic-
6 to 15 hours after the injury. Cytotoxic edema, excitotoxicity, and sec- tive ability of aEEG. Improvement in aEEG tracings may be delayed
ondary energy failure with nearly complete failure of mitochondrial ac- until the patient undergoes rewarming and is no longer sedated.13
tivity characterize this secondary phase, which leads to cell death and Serum biomarkers may enable clinicians to stratify neonates with
clinical deterioration in neonates with moderate to severe injury.6 Sei- HIE undergoing hypothermia into the following 3 groups based on
zures typically occur in the secondary phase.7 A tertiary phase occurs biomarker levels: (1) responders to hypothermia alone with good
during the months after the acute insult and involves late cell death, re- neurodevelopmental prognosis, (2) nonresponders to hypother-
modeling of the injured brain, and astrogliosis.8 mia at high risk for surviving with neurologic injury and/or neurode-
velopmental deficits who then may be candidates for other clinical
interventions, and (3) neonates who will die. Biomarkers, such as
ubiquitin carboxyl-terminal esterase L1 (UCH-L1) and glial fibrillary
Diagnostic Considerations
acidic protein (GFAP), have demonstrated predictive capabilities in
A bedside test is not available for an accurate diagnosis of HIE in a several studies (eTable 2 in the Supplement). Combining biomark-
neonate. Physicians diagnose HIE based on the presence of a neu- ers with scoring systems may improve the sensitivity and specific-

398 JAMA Pediatrics April 2015 Volume 169, Number 4 (Reprinted) jamapediatrics.com

Copyright 2015 American Medical Association. All rights reserved.

Downloaded From: http://archpedi.jamanetwork.com/ by a University of Iowa User on 05/23/2015


Hypoxic-Ischemic Encephalopathy Review Clinical Review & Education

ity of outcome prediction (the Eunice Kennedy Shriver National In- The clinician should be cautious when predicting outcomes in
stitute of Child Health and Human Development HIE calculator is neonates with HIE who have normal findings or minor degrees of
found at http://hopefn3.org). brain injury on MRI. As many as 26% of neonates with HIE who un-
derwent hypothermia and had normal MRI findings experienced ab-
Placental Abnormalities normal neurodevelopmental outcomes.23
Only a small fraction of patients with HIE (15%-29%) have a docu-
mented sentinel event, such as a placental abruption, uterine rup-
ture, cord prolapse, or shoulder dystocia.14,15 In neonates without a
Treatment
sentinel event, placental analysis can provide valuable information
regarding the cause and timing of the adverse events in utero. For Systemic Support
example, placentas with decreased maturation of the terminal villi Systemic support remains the foundation of care for neonates with
are associated with injury to the white matter/watershed areas and HIE. The goal of systemic support is to restore adequate cerebral
basal ganglia.4 Immature placental villi increase the distance be- blood flow, which ensures delivery of the metabolic substrates oxy-
tween the maternal and fetal blood with a net effect of reduced oxy- gen and glucose to prevent secondary brain injury (an overview of
gen diffusion to the fetus or fetal hypoxia. Placentas with reduced recommendations is available in the eFigure in the Supplement). Sec-
weight can represent an adverse intrauterine environment owing ondary injury may occur because of other organ impairment. For ex-
to decreased uteroplacental perfusion.4 ample, cardiac injury may result in decreased cardiac output and hy-
potension, which further decrease cerebral blood flow. Persistent
Neuroimaging pulmonary hypertension of the newborn may worsen hypoxia. Al-
Brain MRI is the preferred imaging choice in neonates with HIE and is though systemic support is the foundation of therapy, evidence-
a useful tool to predict long-term outcomes (eTable 3 in the Supple- based optimal practice parameters are scarce. Researchers have not
ment). In the first week after birth, diffusion-weighted MRI of the brain validated most of the parameters with long-term follow-up of the
mayassistphysiciansinmakingmanagementdecisionsforpatientsun- patients. We herein present a summary of a system-based ap-
dergoingventilatorsupport.16 Diffusion-weightedimagingreferstoMRI proach to supportive care of neonates with HIE.
that is sensitive to water molecule diffusion.17 However, diffusion-
weighted imaging obtained during the first hours after the injury may Respiratory System
underestimate the final extent of injury.16 The sensitivity and specific- Infants with an HI insult have metabolic changes that lead to less car-
ity of this technique can be improved by quantification of the appar- bon dioxide (CO2) production. Respiratory compensation for the ini-
ent diffusion coefficient, which is performed by voxelwise analysis of tial severe metabolic acidosis may lower CO2 levels. In addition, hy-
the information contained within diffusion-weighted imaging.17 After pothermia may reduce CO2 production.24 Patients with HIE need less
moderate or severe HIE, abnormal signal intensity is commonly de- ventilator support to obtain a desirable CO2 level. Hypocapnia is
tected in the basal ganglia and thalami, corticospinal tract, white mat- harmful in patients with HIE because it decreases cerebral perfu-
ter, and cortex.17 Neonates with a history of a sentinel event are likely sion and oxygen release from hemoglobin. Hypocapnia is associ-
to sustain basal ganglia and thalamic lesions.16 These lesions are usu- ated with death and poor neurodevelopmental outcomes.24,25
ally accompanied by abnormalities in the appearance of the interven- Hyperoxia can have a detrimental effect on neonates with HIE
ing posterior limb of the internal capsule.16 because it increases oxidative stress and free radical production, es-
Abnormalities in the MRI of the brain correlate with outcomes. pecially during the reperfusion phase. Furthermore, hyperoxia is as-
Lower apparent diffusion coefficient values in the basal ganglia during sociated with death and poor long-term outcomes in neonates with
the first 7 days after HIE predict adverse neurologic outcomes.18 Inju- HIE.25 Infants with a history of respiratory depression at birth and
ries to the posterior limb of the internal capsule and basal ganglia are resultant HIE often undergo vigorous resuscitation at birth. As a re-
associated with motor deficits.19 Injury to the posterior limb of the in- sult, hyperoxia and hypocapnia may exist after resuscitation, lead-
ternal capsule combined with diffuse basal ganglia injury and a periph- ing to worse outcomes.24,25 Therefore, normal oxygenation and nor-
eral(ie,hemisphericgrayandwhitematter)abnormalityareassociated mocapnia after newborn resuscitation may prevent secondary injury
withdeath,hearingandvisualimpairments,andseverecerebralpalsy.20 (PaCO2, 40-55 mm Hg; PaO2, 50-100 mm Hg).
Recently, the TOBY (Total Body Hypothermia for Neonatal Encepha-
lopathy) trial demonstrated that hypothermia does not influence the Cardiovascular System
ability of MRI to predict neurodevelopmental outcomes.21 Blood pressure must remain in a safe range to avoid hypotension,
Magnetic resonance spectroscopy allows for in vivo quantita- which can produce a secondary ischemic injury. The ideal mean ar-
tive analysis of brain metabolites and therefore may serve as an early terial blood pressure (MAP) for term infants with HIE has not been
biomarker for brain injury. Findings on MRI without spectroscopy established. Because infants operate within a narrow blood pres-
could be normal for as long as 24 hours after an acute HI event (eg, sure range and because HI impairs cerebral autoregulation, ex-
abruption), but magnetic resonance spectroscopy or diffusion- perts recommend that MAP be maintained within the critical range
weighted imaging detects early acute events. When clinicians add of 40 to 60 mm Hg unless the hemodynamics suggest a more op-
magnetic resonance spectroscopy to standard MRI, scanning time timal MAP.26 Organ-specific regional oximetry may indicate the op-
increases by only 6 to 7 minutes and may improve the predictive timal MAP for individual patients, helping to individualize care.27 The
value of the scan. An elevated ratio of lactate to N-acetyl aspartate use of echocardiography in patients with HIE is useful because the
in the basal ganglia can predict long-term neurologic impairments treatment of low pressure is different in infants with poor cardiac
and can be seen in the first 48 hours of life.22 function vs neonates with normal function. Patients with HIE, good

jamapediatrics.com (Reprinted) JAMA Pediatrics April 2015 Volume 169, Number 4 399

Copyright 2015 American Medical Association. All rights reserved.

Downloaded From: http://archpedi.jamanetwork.com/ by a University of Iowa User on 05/23/2015


Clinical Review & Education Review Hypoxic-Ischemic Encephalopathy

cardiac function, and low blood pressure may require more vol- tically with hypothermia, but its efficacy in neonates is unknown.
ume, especially if clinical or historic evidence of hypovolemia (ie, se- Levetiracetam is also a promising antiseizure medication that de-
vere anemia, placental abruption, or cord compression) is found. creases excitotoxicity and does not induce neuronal apoptosis, but
However, the unwarranted use of fluid therapy may exacerbate ce- researchers have not yet evaluated its efficacy in large clinical trials.
rebral edema.28
In patients with cardiac dysfunction, minimal evidence exists re- Hypothermia
garding the ideal method to augment MAP for infants with HIE. A Therapeutic hypothermia is considered the standard of care for neo-
2002 Cochrane systemic review29 did not find conclusive evi- nates with HIE; the treatment uses mild hypothermia in the range
dence regarding the use of dopamine for the prevention of morbid- of 33.5°C to 35.0°C. Several large multicenter trials demonstrated
ity and mortality in patients with HIE. Furthermore, dopamine may that the therapy is safe and efficacious (eTable 4 in the
not be the ideal first-line agent for infants with evidence of pulmo- Supplement).38-40 A recent meta-analysis reviewed outcomes of 7
nary hypertension and HIE because dopamine increases systemic hypothermia trials, including 1214 neonates who were randomized
and pulmonary vascular resistance.30 Dobutamine can reduce af- to hypothermia or systemic supportive care.41 Therapeutic hypo-
terload and therefore decrease the ratio of systemic to pulmonary thermia reduced the risk for death or major neurodevelopmental
vascular resistance.31 Epinephrine at low to moderate doses in- disabilities at 18 months of age in neonates with moderate and se-
creases the cardiac index with no effects on the ratio of systemic to vere HIE.41
pulmonary arterial pressures. In infants with HIE and pulmonary hy- At present, the 2 types of treatment used include whole-body
pertension with cardiac dysfunction, epinephrine may be the opti- hypothermia and selective head cooling. Although the 2 cooling
mal choice for blood pressure augmentation.30 In patients with HIE methods are equally effective, clinicians predominantly use whole-
and pulmonary hypertension, milrinone lactate may be advanta- body cooling owing to its reduced cost and ease of use.
geous because milrinone increases myocardial contractility and acts Meta-analysis41 did not show a difference in the reduction of long-
as a systemic and pulmonary vasodilator.32 term neurologic impairments between the 2 methods.
Recently, 2 of the original large multicenter hypothermia trials
Fluids, Electrolytes, and Nutrition published follow-up data on their original cohort of patients at school
Research has not been conducted on the optimal initial rate of fluid age.42,43 The CoolCap Trial Group42 performed neurodevelopmen-
therapy and its long-term outcomes. To limit the consequences of tal assessments on 46% of the original cohort and demonstrated a
HIE, experts recommend carefully managing fluid therapy to avoid correlation between the neurodevelopmental assessments at 18 to
fluid overload and thus prevent cerebral edema.33 Recommenda- 22 months of age with the functional outcomes at 7 to 8 years of
tions for neonatal fluid restriction are based on the experience of age. The Eunice Kennedy Shriver National Institute of Child Health
restricting fluid intake in adults and older children with cerebral and Human Development Neonatal Research Network whole-
edema to a target of 40 to 70 mL/kg/d.33 body hypothermia trial43 demonstrated a significant reduction in
In normal conditions, the human brain relies almost entirely on death, death or severe disability, and death or cerebral palsy at 6 to
glucose to provide the substrate for metabolism.34 Neonatal cere- 7 years of age. The trial also demonstrated a strong trend in the pri-
bral glucose consumption may account for 70% of total glucose mary outcome of death or IQ score of less than 70.43
consumption.34 Although the newborn brain can use other sub- Because little variability was evident in the therapeutic hypo-
strates (ie, lactate or ketones) as an energy source, these alternate thermia trials, the optimal timing for the initiation of hypothermia,
substrates have an unpredictable supply and may not compensate the depth and duration of hypothermia, and the safety of hypother-
entirely for a decrease in glucose availability.34 During HI, anaero- mia for late preterm neonates are uncertain. These uncertainties are
bic glycolysis depletes hepatic glycogen stores, and hepatic glu- being addressed in ongoing trials that examine the efficacy of ini-
cose production rapidly becomes insufficient to meet cerebral meta- tiation of hypothermia at 6 to 24 hours of age,44 longer durations
bolic demands.34 Clinical observations demonstrate a correlation of hypothermia (120 hours) and a lower temperature of hypother-
between lower serum glucose concentrations and higher neonatal mia (32°C),45 and whether neonates with HIE and a gestational age
Sarnat stages.35 In addition, initial hypoglycemia (ⱕ40 mg/dL [to of 33 to 35 weeks benefit from hypothermia.46
convert to millimoles per liter, multiply by 0.0555]) is an important Recent data showed that the time to initiate hypothermia cor-
risk factor for perinatal brain injury in neonates with HI.36 There- related with outcomes. Neonates undergoing earlier cooling therapy
fore, strict monitoring of glucose levels is necessary to prevent and (within 180 minutes of birth) had better outcomes compared with
treat hypoglycemia. Fluid restrictions can compromise appropri- those who underwent the therapy later (180-360 minutes after
ate glucose delivery and may contribute to hypoglycemia. birth).47 The results have stimulated interest in transporting neo-
nates with active hypothermia.48-50
Antiseizure Medications
Consensus has not been reached regarding the best medication for Emerging Therapies
treating seizures in patients with HIE. Clinicians frequently use phe- Although the pathophysiological features of HIE are complex, the
nobarbital, but only 27% of seizures are controlled. Recently, topi- multiple steps leading to cellular damage provide many opportuni-
ramate has emerged as a potential neonatal antiseizure medica- ties for therapeutic intervention. A search is currently under way to
tion. Topiramate modulates 2-(aminomethyl)phenylacetic acid, identify other agents that may be synergistic with therapeutic hy-
kainate, and γ-aminobutyric acid–activated ion channels and voltage- pothermia. Potential agents include xenon, erythropoietin, mela-
activated sodium and chloride channels. Animal models and 1 hu- tonin, and stem cell therapy. A brief discussion of these agents fol-
man pilot clinical trial37 showed that topiramate worked synergis- lows. These agents, along with other potential therapies, are

400 JAMA Pediatrics April 2015 Volume 169, Number 4 (Reprinted) jamapediatrics.com

Copyright 2015 American Medical Association. All rights reserved.

Downloaded From: http://archpedi.jamanetwork.com/ by a University of Iowa User on 05/23/2015


Hypoxic-Ischemic Encephalopathy Review Clinical Review & Education

Figure 2. Complex Cascade of Events Producing Cellular Damage and Destruction After Hypoxia-Ischemia (HI)

Axon Dendrite
Neurosynaptic Junction

Na+ Membrane depolarization


Xanthine

Free radicals 8a

Uric acid
KA-AMPA Angiogenesis
5 9 3
Lipases Lipid peroxidation
HI Neurogenesis
Ca2+ Nucleases
Glutamate Glutamate Cell membrane
destruction
NO synthase

Degradation
Free radicals
of
NMDA microtubules Free radicals 9
2

6 8 10
6 8 10
1
3 4 5 10
Ca2+

7
Apoptosis/necrosis

During hypoxic-ischemic encephalopathy, an excessive amount of the neuroprotective agents are illustrated at the points where they interfere with
excitatory amino acid glutamate is released from the presynaptic terminal. This the pathophysiological cascade. Solid arrows represent the pathophysiological
excess glutamate leads to overstimulation of the glutamate receptors cascade that is unleashed as a result of HI injury; dashed arrows, interruption of
(2-(aminomethyl)phenylacetic acid [AMPA], kainite [KA], and the cascade by the various neuroprotective agents (circled numbers). As in
N-methyl-D-aspartate [NMDA]) located on the postsynaptic neuron and leads to Figure 1, the orange boxes represent excitotoxicity; blue boxes, oxidative
excitotoxicity. Overstimulation of the KA and AMPA receptors causes sodium stress; yellow box, repair; light green box, cell death; and dark blue box, HI.
(Na+) and chloride to enter the cell, which increases cell osmolality. NO indicates nitric oxide; 1, magnesium; 2, xenon; 3, erythropoietin; 4, stem
Overstimulation of the NMDA receptor triggers the influx of calcium (Ca2+). cells; 5, N-acetylcysteine; 6, melatonin; 7, anticonvulsants; 8, antioxidants;
The 3 aberrant cellular processes lead to apoptosis and necrosis. The various 8a, allopurinol sodium; 9, BH4 (tetrahydrobiopterin); and 10, hydrogen sulfide.

summarized in Figure 2. More extensive reviews on emerging neu- inflammatory effect when bound to erythropoietin receptors on
roprotective therapies are available.51 astrocytes and microglial cells.56 Erythropoietin prevents nitric oxi-
Because excitatory amino acids play an important role in the cas- de–induced death of neurons and protects neurons from the toxic
cade of events leading to cell death, researchers have identified phar- effects of glutamate.57 Erythropoietin is neurotrophic and affects
macologic agents that inhibit excitatory amino acid release or block neurogenesis, differentiation, and repair after injury.58 In the study
their postsynaptic actions. Xenon, a noble gas currently used as an by Zhu et al,59 neonates with moderate or severe HIE were random-
inhaled anesthetic, is an N-methyl-D-aspartate receptor antago- ized to receive erythropoietin or supportive care without hypother-
nist. Xenon may have other neuroprotective qualities, such as af- mia therapy. Erythropoietin was administered within 48 hours of
fecting other ion channels and reducing neurotransmitter release in birth at a dose of 300 U/kg or 500 U/kg every other day for 2 weeks.
general. Xenon is an attractive agent for use in infants with HIE be- At 18 months of age, the erythropoietin group had reduced rates of
cause it easily crosses the blood-brain barrier, takes rapid effect, can death and moderate or severe disability. The outcome was the same
be rapidly withdrawn, and has myocardial protective properties with when the 300- and 500-U/kg doses were compared. Researchers
very limited potential cardiovascular effects.52 A phase 1 trial using saw these improvements only in patients with moderate, not se-
xenon in combination with hypothermia was completed recently and vere, HIE. Recently, a phase 1 pharmacokinetic study combined eryth-
demonstrated xenon administration to be feasible with no adverse ropoietin with hypothermia and demonstrated that participants tol-
cardiovascular or respiratory effects.53 A phase 2 study is currently erated doses up to 1000 U/kg. This dose produced plasma
under way. concentrations similar to those in animal models of HI injury that are
Erythropoietin is a naturally occurring glycoprotein frequently neuroprotective.60
used to stimulate erythropoiesis and is a safe and efficacious treat- The pineal gland produces melatonin, a naturally occurring sub-
ment for anemia of prematurity.54 Erythropoietin is locally pro- stance used for regulating the circadian rhythm. Melatonin has many
duced in the central nervous system and is found in elevated levels other effects that may benefit infants with HI injury. Melatonin serves
in the cord blood of infants who have perinatal asphyxia.55 Eryth- as a free radical scavenger of the hydroxyl radical, oxygen, and hy-
ropoietin has many possible mechanisms for neuroprotection. It pro- drogen peroxide. In addition, melatonin decreases inflammatory
vides neuroprotection against apoptosis and has an anti- cytokine levels and stimulates antioxidant enzymes, such as gluta-

jamapediatrics.com (Reprinted) JAMA Pediatrics April 2015 Volume 169, Number 4 401

Copyright 2015 American Medical Association. All rights reserved.

Downloaded From: http://archpedi.jamanetwork.com/ by a University of Iowa User on 05/23/2015


Clinical Review & Education Review Hypoxic-Ischemic Encephalopathy

thione peroxidase and reductase, glucose-6-phosphate dehydro- alone.63 Validation of the efficacy of stem cell therapy will require a
genase, and superoxide dismutase.61 In a cohort study of new- bigger sample size and protocols that standardize the source of cells,
borns with asphyxia, melatonin (10 mg by mouth given every 2 hours doses, and method of delivery.
for 8 doses) reduced malondialdehyde and nitrate/nitrite levels com-
pared with placebo.62 These reduced levels demonstrated a de-
crease in lipid peroxidation and nitric oxide production.
Conclusions
Stem cell therapy may be a good adjunctive therapy because
of its potential for benefit through several different mechanisms. Great strides in the care of neonates with HIE have been made. The
Stem cell transplant may increase levels of brain trophic factors and advent of therapeutic hypothermia has armed the bedside clinician
antiapoptotic factors, decrease inflammation, preserve endoge- with a therapy that helps reduce the long-term neurologic impair-
nous tissue, and support replacement of damaged cells.8 A recent ments in 1 of 8 treated neonates. However, the fields of neonatology
pilot study showed better outcomes with the combination of hy- and neonatal neurology should continue to search for neuroprotec-
pothermia plus autologous umbilical cord blood vs hypothermia tive strategies and long-term optimization of infant neuroplasticity.

ARTICLE INFORMATION prolonged selective head cooling after ischemia in 20. Jyoti R, O’Neil R, Hurrion E. Predicting
Accepted for Publication: November 2, 2014. fetal lambs. J Clin Invest. 1997;99(2):248-256. outcome in term neonates with hypoxic-ischaemic
8. Bennet L, Tan S, Van den Heuij L, et al. encephalopathy using simplified MR criteria.
Published Online: February 16, 2015. Pediatr Radiol. 2006;36(1):38-42.
doi:10.1001/jamapediatrics.2014.3269. Cell therapy for neonatal hypoxia-ischemia and
cerebral palsy. Ann Neurol. 2012;71(5):589-600. 21. Rutherford M, Ramenghi LA, Edwards AD, et al.
Author Contributions: Drs Douglas-Escobar and Assessment of brain tissue injury after moderate
Weiss had full access to all the data in the study and 9. Volpe JJ. Neonatal encephalopathy:
an inadequate term for hypoxic-ischemic hypothermia in neonates with hypoxic-ischaemic
take responsibility for the integrity of the data and encephalopathy: a nested substudy of a
the accuracy of the data analysis. encephalopathy. Ann Neurol. 2012;72(2):156-166.
randomised controlled trial. Lancet Neurol. 2010;9
Study concept and design: All authors. 10. Güneś T, Oztürk MA, Köklü SM, Narin N, (1):39-45.
Acquisition, analysis, or interpretation of data: Köklü E. Troponin-T levels in perinatally asphyxiated
Douglas-Escobar. infants during the first 15 days of life. Acta Paediatr. 22. Heinz ER, Provenzale JM. Imaging findings in
Drafting of the manuscript: All authors. 2005;94(11):1638-1643. neonatal hypoxia: a practical review. AJR Am J
Critical revision of the manuscript for important Roentgenol. 2009;192(1):41-47.
11. Sarnat HB, Sarnat MS. Neonatal encephalopathy
intellectual content: All authors. following fetal distress: a clinical and 23. Rollins N, Booth T, Morriss MC, Sanchez P,
Study supervision: All authors. electroencephalographic study. Arch Neurol. 1976; Heyne R, Chalak L. Predictive value of neonatal MRI
Conflict of Interest Disclosures: None reported. 33(10):696-705. showing no or minor degrees of brain injury after
hypothermia. Pediatr Neurol. 2014;50(5):447-451.
Additional Contributions: Roger Hoover (News 12. van Laerhoven H, de Haan TR, Offringa M,
and Public Affairs, University of Florida) provided Post B, van der Lee JH. Prognostic tests in term 24. Pappas A, Shankaran S, Laptook AR, et al;
the artwork for Figures 1 and 2. He received no neonates with hypoxic-ischemic encephalopathy: Eunice Kennedy Shriver National Institute of Child
financial compensation. a systematic review. Pediatrics. 2013;131(1):88-98. Health and Human Development Neonatal
Research Network. Hypocarbia and adverse
13. Thoresen M, Hellström-Westas L, Liu X, outcome in neonatal hypoxic-ischemic
REFERENCES de Vries LS. Effect of hypothermia on encephalopathy. J Pediatr. 2011;158(5):752-758.e1.
1. Kurinczuk JJ, White-Koning M, Badawi N. amplitude-integrated electroencephalogram in doi:10.1016/j.jpeds.2010.10.019.
Epidemiology of neonatal encephalopathy and infants with asphyxia. Pediatrics. 2010;126(1):
hypoxic-ischaemic encephalopathy. Early Hum Dev. e131-e139. doi:10.1542/peds.2009-2938. 25. Klinger G, Beyene J, Shah P, Perlman M.
2010;86(6):329-338. Do hyperoxaemia and hypocapnia add to the risk of
14. Badawi N, Kurinczuk JJ, Keogh JM, et al. brain injury after intrapartum asphyxia? Arch Dis
2. Johnston MV, Fatemi A, Wilson MA, Intrapartum risk factors for newborn Child Fetal Neonatal Ed. 2005;90(1):F49-F52.
Northington F. Treatment advances in neonatal encephalopathy: the Western Australian
neuroprotection and neurointensive care. Lancet case-control study. BMJ. 1998;317(7172):1554-1558. 26. Volpe JJ. Neurology of the Newborn. Philadelphia,
Neurol. 2011;10(4):372-382. PA: WB Saunders; 2008.
15. Nelson KB, Bingham P, Edwards EM, et al.
3. Ferriero DM. Neonatal brain injury. N Engl J Med. Antecedents of neonatal encephalopathy in the 27. Howlett JA, Northington FJ, Gilmore MM, et al.
2004;351(19):1985-1995. Vermont Oxford Network Encephalopathy Registry. Cerebrovascular autoregulation and neurologic
Pediatrics. 2012;130(5):878-886. injury in neonatal hypoxic-ischemic
4. Harteman JC, Nikkels PG, Benders MJ, Kwee A, encephalopathy. Pediatr Res. 2013;74(5):525-535.
Groenendaal F, de Vries LS. Placental pathology in 16. Rutherford M, Biarge MM, Allsop J, Counsell S,
full-term infants with hypoxic-ischemic neonatal Cowan F. MRI of perinatal brain injury. Pediatr Radiol. 28. Kimelberg HK. Current concepts of brain
encephalopathy and association with magnetic 2010;40(6):819-833. edema: review of laboratory investigations.
resonance imaging pattern of brain injury. J Pediatr. J Neurosurg. 1995;83(6):1051-1059.
17. Azzopardi D, Edwards AD. Magnetic resonance
2013;163(4):968-95.e2. doi:10.1016/j.jpeds.2013. biomarkers of neuroprotective effects in infants 29. Hunt R, Osborn D. Dopamine for prevention of
06.010. with hypoxic ischemic encephalopathy. Semin Fetal morbidity and mortality in term newborn infants
5. Wassink G, Gunn ER, Drury PP, Bennet L, Gunn Neonatal Med. 2010;15(5):261-269. with suspected perinatal asphyxia. Cochrane
AJ. The mechanisms and treatment of asphyxial Database Syst Rev. 2002;(3):CD003484.
18. Barkovich AJ, Miller SP, Bartha A, et al.
encephalopathy. Front Neurosci. 2014;8:40. MR imaging, MR spectroscopy, and diffusion tensor 30. Cheung PY, Barrington KJ. The effects of
doi:10.3389/fnins.2014.00040. imaging of sequential studies in neonates with dopamine and epinephrine on hemodynamics and
6. Bennet L, Roelfsema V, Pathipati P, encephalopathy. AJNR Am J Neuroradiol. 2006;27 oxygen metabolism in hypoxic anesthetized piglets.
Quaedackers JS, Gunn AJ. Relationship between (3):533-547. Crit Care. 2001;5(3):158-166.
evolving epileptiform activity and delayed loss of 19. Hunt RW, Neil JJ, Coleman LT, Kean MJ, 31. Cheung PY, Barrington KJ, Bigam D.
mitochondrial activity after asphyxia measured by Inder TE. Apparent diffusion coefficient in the The hemodynamic effects of dobutamine infusion
near-infrared spectroscopy in preterm fetal sheep. posterior limb of the internal capsule predicts in the chronically instrumented newborn piglet. Crit
J Physiol. 2006;572(pt 1):141-154. outcome after perinatal asphyxia. Pediatrics. 2004; Care Med. 1999;27(3):558-564.
7. Gunn AJ, Gunn TR, de Haan HH, Williams CE, 114(4):999-1003. 32. McNamara PJ, Laique F, Muang-In S, Whyte HE.
Gluckman PD. Dramatic neuronal rescue with Milrinone improves oxygenation in neonates with

402 JAMA Pediatrics April 2015 Volume 169, Number 4 (Reprinted) jamapediatrics.com

Copyright 2015 American Medical Association. All rights reserved.

Downloaded From: http://archpedi.jamanetwork.com/ by a University of Iowa User on 05/23/2015


Hypoxic-Ischemic Encephalopathy Review Clinical Review & Education

severe persistent pulmonary hypertension of the Network. Childhood outcomes after hypothermia 54. Maier RF, Obladen M, Scigalla P, et al; European
newborn. J Crit Care. 2006;21(2):217-222. for neonatal encephalopathy. N Engl J Med. 2012; Multicentre Erythropoietin Study Group. The effect
33. Kecskes Z, Healy G, Jensen A. Fluid restriction 366(22):2085-2092. of epoetin beta (recombinant human
for term infants with hypoxic-ischaemic 44. ClinicalTrials.gov. Late Hypothermia for erythropoietin) on the need for transfusion in
encephalopathy following perinatal asphyxia. Hypoxic-Ischemic Encephalopathy. NCT00614744. very-low-birth-weight infants. N Engl J Med. 1994;
Cochrane Database Syst Rev. 2005;(3):CD004337. https://clinicaltrials.gov/ct2/show/NCT00614744. 330(17):1173-1178.

34. McGowan JE, Perlman JM. Glucose Accessed January 22, 2015. 55. Ruth V, Widness JA, Clemons G, Raivio KO.
management during and after intensive delivery 45. ClinicalTrials.gov. Optimizing (Longer, Deeper) Postnatal changes in serum immunoreactive
room resuscitation. Clin Perinatol. Cooling for Neonatal Hypoxic-Ischemic erythropoietin in relation to hypoxia before and
2006;33(1):183-196, x. Encephalopathy (HIE). NCT01192776. after birth. J Pediatr. 1990;116(6):950-954.

35. Basu P, Som S, Choudhuri N, Das H. https://clinicaltrials.gov/ct2/show/NCT01192776. 56. Sun Y, Calvert JW, Zhang JH. Neonatal
Contribution of the blood glucose level in perinatal Accessed January 22, 2015. hypoxia/ischemia is associated with decreased
asphyxia. Eur J Pediatr. 2009;168(7):833-838. 46. ClinicalTrials.gov. Preemie Hypothermia for inflammatory mediators after erythropoietin
Neonatal Encephalopathy. NCT01793129. administration. Stroke. 2005;36(8):1672-1678.
36. Salhab WA, Wyckoff MH, Laptook AR,
Perlman JM. Initial hypoglycemia and neonatal https://clinicaltrials.gov/ct2/show/NCT01793129. 57. Sakanaka M, Wen TC, Matsuda S, et al. In vivo
brain injury in term infants with severe fetal Accessed January 22, 2015. evidence that erythropoietin protects neurons from
acidemia. Pediatrics. 2004;114(2):361-366. 47. Thoresen M, Tooley J, Liu X, et al. Time is brain: ischemic damage. Proc Natl Acad Sci U S A. 1998;95
starting therapeutic hypothermia within three (8):4635-4640.
37. Filippi L, la Marca G, Fiorini P, et al. Topiramate
concentrations in neonates treated with prolonged hours after birth improves motor outcome in 58. Wang L, Zhang Z, Zhang R, et al. Erythropoietin
whole body hypothermia for hypoxic ischemic asphyxiated newborns. Neonatology. 2013;104(3): up-regulates SOCS2 in neuronal progenitor cells
encephalopathy. Epilepsia. 2009;50(11):2355-2361. 228-233. derived from SVZ of adult rat. Neuroreport. 2004;15
48. Hobson A, Sussman C, Knight J, et al. Active (8):1225-1229.
38. Azzopardi DV, Strohm B, Edwards AD, et al;
TOBY Study Group. Moderate hypothermia to treat cooling during transport of neonates with 59. Zhu C, Kang W, Xu F, et al. Erythropoietin
perinatal asphyxial encephalopathy. N Engl J Med. hypoxic-ischemic encephalopathy. Air Med J. 2011; improved neurologic outcomes in newborns with
2009;361(14):1349-1358. 30(4):197-200. hypoxic-ischemic encephalopathy. Pediatrics.
49. Johnston ED, Becher JC, Mitchell AP, Stenson 2009;124(2):e218-e226. doi:10.1542/peds
39. Gunn AJ, Wyatt JS, Whitelaw A, et al; CoolCap .2008-3553.
Study Group. Therapeutic hypothermia changes the BJ. Provision of servo-controlled cooling during
prognostic value of clinical evaluation of neonatal neonatal transport. Arch Dis Child Fetal Neonatal Ed. 60. Wu YW, Bauer LA, Ballard RA, et al.
encephalopathy. J Pediatr. 2008;152(1):55-58, 58.e1. 2012;97(5):F365-F367. Erythropoietin for neuroprotection in neonatal
doi:10.1016/j.jpeds.2007.06.003. 50. O’Reilly KM, Tooley J, Winterbottom S. encephalopathy: safety and pharmacokinetics.
Therapeutic hypothermia during neonatal Pediatrics. 2012;130(4):683-691.
40. Shankaran S, Laptook AR, Ehrenkranz RA, et al;
National Institute of Child Health and Human transport. Acta Paediatr. 2011;100(8):1084-1086. 61. Lee MY, Kuan YH, Chen HY, et al. Intravenous
Development Neonatal Research Network. doi:10.1111/j.1651-2227.2011.02249.x. administration of melatonin reduces the
Whole-body hypothermia for neonates with 51. Hobson A, Baines J, Weiss MD. Beyond intracerebral cellular inflammatory response
hypoxic-ischemic encephalopathy. N Engl J Med. hypothermia: alternative therapies for following transient focal cerebral ischemia in rats.
2005;353(15):1574-1584. hypoxic-ischemic encephalopathy. Open J Pineal Res. 2007;42(3):297-309.

41. Tagin MA, Woolcott CG, Vincer MJ, Whyte RK, Pharmacol J. 2013;7:26-40. 62. Fulia F, Gitto E, Cuzzocrea S, et al. Increased
Stinson DA. Hypothermia for neonatal hypoxic 52. David HN, Haelewyn B, Rouillon C, et al. levels of malondialdehyde and nitrite/nitrate in the
ischemic encephalopathy: an updated systematic Neuroprotective effects of xenon: a therapeutic blood of asphyxiated newborns: reduction by
review and meta-analysis. Arch Pediatr Adolesc Med. window of opportunity in rats subjected to melatonin. J Pineal Res. 2001;31(4):343-349.
2012;166(6):558-566. transient cerebral ischemia. FASEB J. 2008;22(4): 63. Cotten CM, Murtha AP, Goldberg RN, et al.
42. Guillet R, Edwards AD, Thoresen M, et al; 1275-1286. Feasibility of autologous cord blood cells for infants
CoolCap Trial Group. Seven- to eight-year follow-up 53. Dingley J, Tooley J, Liu X, et al. Xenon with hypoxic-ischemic encephalopathy. J Pediatr.
of the CoolCap trial of head cooling for neonatal ventilation during therapeutic hypothermia in 2014;164(5):973-979.e1. doi:10.1016/j.jpeds.2013.11
encephalopathy. Pediatr Res. 2012;71(2):205-209. neonatal encephalopathy: a feasibility study. .036.

43. Shankaran S, Pappas A, McDonald SA, et al; Pediatrics. 2014;133(5):809-818.


Eunice Kennedy Shriver NICHD Neonatal Research

jamapediatrics.com (Reprinted) JAMA Pediatrics April 2015 Volume 169, Number 4 403

Copyright 2015 American Medical Association. All rights reserved.

Downloaded From: http://archpedi.jamanetwork.com/ by a University of Iowa User on 05/23/2015

You might also like