Pearlman 2004

download Pearlman 2004

of 17

Transcript of Pearlman 2004

  • 7/30/2019 Pearlman 2004

    1/17

    Hypoxicischemic brain injury in the term

    infant-current concepts

    Lina Shalaka, Jeffrey M. Perlmanb,*

    aDepartment of Pediatrics, Southwestern Medical Center, University of Texas, 5323 Harry Hines Blvd.,

    75390 Dallas, TX, United StatesbDepartment of Pediatrics, Weill Medical College of Cornell University, New York, NY 10021, United States

    Accepted 4 June 2004

    Abstract

    An enhanced understanding of the cellular characteristics contributing to ongoing brain injury

    following intrapartum hypoxia-ischemia has resulted in the implementation of targeted neuro-

    protective strategies in the newborn period. This review briefly covers the pathogenesis of hypoxic-

    ischemic injury with an emphasis on reperfusion injury; the role of magnetic resonance imaging in

    the detection of such injury, and focuses on potential strategies both supportive and neuroprotectiveto prevent ongoing injury with a specific emphasis on modest hypothermia.

    D 2004 Elsevier Ireland Ltd. All rights reserved.

    Keywords: Hypoxicischemic brain injury; Infant-current concepts cerebral palsy

    1. Background

    Hypoxicischemic cerebral injury that occurs during the perinatal period is one of the

    most commonly recognized causes of severe, long-term neurological deficits in children,

    often referred to as cerebral palsy (CP) [1,2]. The brain injury that develops is an evolvingprocess initiated during the insult and extends into a recovery period, the latter referred to as

    the breperfusion phaseQ of injury. Clinically, it is the latter phase that is amendable to

    potential intervention(s). Until recently, current management strategies have largely been

    0378-3782/$ - see front matterD 2004 Elsevier Ireland Ltd. All rights reserved.

    doi:10.1016/j.earlhumdev.2004.06.003

    * Corresponding author.

    Early Human Development 80 (2004) 125141www.elsevier.com/locate/earlhumdev

  • 7/30/2019 Pearlman 2004

    2/17

    supportive and not targeted toward the processes of ongoing injury [3]. Thus, it should not be

    surprising that, despite improvements in perinatal practice during the past several decades,

    the incidence of CP attributed to intrapartum asphyxia has remained essentially unchanged[1]. However, novel exciting strategies aimed at preventing ongoing injury are being

    clinically evaluated and offer an opportunity for neuroprotection. In this discussion, we

    briefly review the cellular characteristics of hypoxicischemic cerebral injury and the

    current and future therapeutic strategies aimed at ameliorating ongoing brain injury

    following intrapartum hypoxiaischemia.

    2. Pathogenesis of hypoxicischemic cerebral injury

    The principal pathogenetic mechanism underlying most of the neuropathology

    attributed to intrapartum hypoxiaischemia is impaired cerebral blood flow (CBF). Thisis most likely to occur as a consequence of interruption in placental blood flow and gas

    exchange, often referred to as basphyxiaQ or severe fetal acidemia. The latter is defined as a

    fetal umbilical arterial pHV7.00 [1].

    At the cellular level, the reduction in cerebral blood flow and oxygen delivery initiates a

    cascade of deleterious biochemical events. Depletion of oxygen precludes oxidative

    phosphorylation and results in a switch to anaerobic metabolism. This is an energy

    inefficient state resulting in rapid depletion of high-energy phosphate reserves including

    ATP, accumulation of lactic acid and the inability to maintain cellular functions [4].

    Transcellular ion pump failure results in the intracellular accumulation of Na+, Ca+ and water

    (cytotoxic edema). The membrane depolarization results in a release of excitatory

    neurotransmitters and specifically glutamate from axon terminals. The glutamate then

    activates specific cell surface receptors resulting in an influx of Na+ and Ca+ into

    Fig. 1. Potential biochemical mechanisms of hypoxic ischemic cerebral injury.

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141126

  • 7/30/2019 Pearlman 2004

    3/17

    postsynaptic neurons. Within the cytoplasm, there is an accumulation of free fatty acids

    secondary to increased membrane phospholipid turnover. The fatty acids undergo

    peroxidation by oxygen free radicals that arise from reductive processes within mitochondriaand as byproducts in the synthesis of prostaglandins, xanthine and uric acid (see Fig. 1). Ca+

    ions accumulate within the cytoplasm as a consequence of increased cellular influx as well as

    decreased efflux across the plasma membrane combined with release from mitochondria and

    endoplasmic reticulum. In selected neurons, the intracellular calcium induces the production

    of nitric oxide (NO), a free radical that diffuses to adjacent cells susceptible to nitric oxide

    toxicity. The combined effects of cellular energy failure, acidosis, glutamate release,

    intracellular Ca+ accumulation, lipid peroxidation and nitric oxide neurotoxicity serve to

    disrupt essential components of the cell with its ultimate death [2,5]. Many factors including

    the duration or severity of the insult will influence the progression of cellular injury

    following hypoxiaischemia (Table 1).

    3. Delayed (secondary) brain damage

    Following resuscitation, which may occur in utero or postnatally in the delivery room,

    cerebral oxygenation and perfusion is restored. During this recovery phase, the concen-

    trations of phosphorus metabolites and the intracellular pH return to baseline. However, the

    process of cerebral energy failure recurs from 6 to 48 h later in a second phase of injury. This

    phase is characterized by a decrease in the ratio of phosphocreatine/inorganic phosphate,

    with an unchanged intracellular pH, stable cardiorespiratory status and contributes to further

    brain injury [4,6]. In the human infant, the severity of the second energy failure is correlated

    with adverse neurodevelopmental outcome at 1 and 4 years [7]. The mechanisms of

    secondary energy failure may involve mitochondrial dysfunction secondary to extendedreactions from primary insults (e.g., calcium influx, excitatory neurotoxicity, oxygen free

    radicals or nitric oxide formation). Indeed, mitochondria play a key role in determining the

    fate of neurons following hypoxiaischemia. Thus, translocation of apoptotic triggering

    proteins such as cytochrome c from the mitochondria to the cytoplasm can activate a cascade

    of proteolytic enzymes termed caspases or cysteine proteases that eventually trigger nuclear

    fragmentation [5]. Recent evidence also suggests that circulatory and endogenous

    inflammatory cells/mediators also contribute to ongoing brain injury [8].

    To discuss this process further, four basic mechanisms of ongoing injury are outlined

    next.

    3.1. Accumulation of cytosolic calcium

    Calcium is an intracellular second messenger necessarily for numerous cellular reactions.

    Under physiologic condition, cytosolic Ca+ concentration is strictly regulated at a very low

    intracellular concentration. During hypoxiaischemia, there is an increase in Ca+ influx into

    neuronal cells in part by stimulation of the NMDA receptors of the agonist-operated Ca+

    channel by glutamate. In an opposite direction, Ca+ efflux across the plasma membrane is

    interrupted by energy failure. Moreover, Ca+ is also released into cytoplasm from the

    mitochondria by the Na+,H+-dependent antiport system and from the endoplasmic reticulum

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141 127

  • 7/30/2019 Pearlman 2004

    4/17

    by inositol triphosphate (IP3) derived from the plasma membrane. These alterations of Ca+

    balance result in increased intracellular Ca+, which interferes with many enzymatic reactions

    including the activation of lipases, proteases, endonucleases and phospholipases and theformation of oxygen free radicals as byproducts of xanthine and prostaglandin synthesis.

    Overall, the accumulation of cytosolic Ca+ after hypoxiaischemia has detrimental effects

    on neuronal cells leading to irreversible brain damage [2,5].

    3.2. Excitatory neurotransmitter release

    Glutamate is a major excitatory amino acid within brain. The action of glutamate is

    mediated by a number of receptor subtypes with the NMDA receptor predominating in

    developing brain and is increased in areas of active development, e.g., striatum or

    hippocampus. Within the NMDA receptor site, there are regions of agonist activity, i.e.,

    glutamate, glycine as well as regions of antagonist activity, i.e., Mg

    2+

    , phencyclidine or PCP[2,5]. Each of these latter sites may offer the potential for neuroprotective strategies.

    During hypoxiaischemia, glutamate accumulates within the synaptic cleft secondary to

    increased release from axon terminals as well as impaired reuptake at presynaptic nerve

    endings. The excessive glutamate acting on the receptor sites facilitates the intracellular

    entry of Ca+ via the NMDA receptor-mediated channel and promotes the biochemical

    cascade mentioned above leading to ultimate neuronal death [2,5].

    3.3. Formation of free radicals

    3.3.1. Oxygen free radicals

    In aerobic cells, oxygen free radicals (i.e., O2, H2O2, OH

    ) are produced within the

    cytoplasm and mitochondria. Under physiologic condition, oxygen free radicals aredestroyed rapidly by endogenous antioxidants (i.e., superoxide dismutase, endoperoxidase,

    catalase) and scavengers (i.e., cholesterol, a-tocopherol, ascorbic acid, glutathione) [5,8].

    During and following hypoxiaischemia, the generation of oxygen free radicals is increased

    in excess of the protective capability. Two major sources of oxygen free radicals are the

    byproducts of xanthine (derived from the breakdown of ATP) and prostaglandin synthesis

    (derived from the breakdown of free fatty acids) (see Fig. 1). Oxygen free radicals contribute

    to tissue injury by attacking the polyunsaturated fatty acid component of the cellular

    membrane resulting in membrane fragmentation and cell death [5,8].

    3.3.2. Nitric oxide

    Nitric oxide (NO) is a weak free radical formed during the conversion ofl-arginine to l-citrulline by nitric oxide synthase (NOS). NOS is strongly activated during hypoxia

    ischemia as well as during reperfusion and a large amount of NO is produced for extended

    periods. NO when combined with superoxide generates a potent radical, peroxynitrite,

    which activates lipid peroxidation. In addition, NO also enhances glutamate release [8].

    3.3.3. Iron

    Physiologically, iron (Fe) is maintained in non-toxic ferric state and bound to proteins

    (i.e., ferritin, transferrin). During hypoxiaischemia, free ferric iron is released from these

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141128

  • 7/30/2019 Pearlman 2004

    5/17

    proteins and reacts with peroxides to generate potent hydroxyl radicals. In addition, free

    ferric iron is reduced to a ferrous form, which further contributes to free radical injury [8].

    3.4. Inflammatory mediators

    Inflammatory mediators seem to play a critical role in the pathogenesis of hypoxic

    ischemic brain injury. Thus expression of IL-1-h and TNF a messenger RNA has been

    demonstrated within 14 h following hypoxia ischemia [9], along with the induction ofa

    and h chemokines followed by neutrophil invasion of the area of infarction [10].

    Furthermore, a low dose of bacterial endotoxin administered 4 h prior to hypoxiaischemia

    in rats induces cerebral infarction in response to short periods of hypoxiaischemia that

    usually causes little or no injury. This endotoxin induced sensitization of the immature brain

    occurs independent of cerebral blood flow changes and/or hyperthermia and is associated

    with an altered expression of CD14 mRNA [11]. Inflammatory cytokines may have a directtoxic effect via increased production of inducible nitric oxide synthase, cyclooxygenase and

    free radical release, or indirectly via induction of glial cells to produce neurotoxic factors

    such as excitatory amino acids. The important role of cytokines in perpetuating excitotoxic

    injury is suggested from experimental observations in which administration of IL-1

    endogenous receptor antagonists as well as neutralizing antibody are associated with a

    reduction in excitotoxic and ischemic injury in mice [12,13]. However, cytokines that

    potentiate brain damage following ischemia also seem to play a beneficial neurotrophic

    effect. Thus, TNF knockout mice were shown to have increased neuronal cell degeneration

    following ischemia and excitotoxic injury suggesting a critical role for endogenous TNF in

    regulating the cellular responses to brain injury [14]. In addition, administration of

    exogenous IL-6 following ischemia produced by middle artery occlusion also results in

    marked neuroprotection [15]. Thus, inflammatory cytokines appear to exert both beneficialand deleterious effects following ischemia. This dual effect will likely complicate the task of

    developing targeted interventions against the inflammatory response.

    4. Mechanisms of neuronal cell death following hypoxiaischemia

    The mechanism of neuronal cell death in animals and human following hypoxia

    ischemia includes neuronal necrosis and apoptosis. The intensity of the initial insult may

    determine the mode of death with severe injury resulting in necrosis, while milder insults

    result in apoptosis [16].Necrosis is a passive process of cell swelling, disrupted cytoplasmic

    organelles, loss of membrane integrity and eventual lysis of neuronal cells and activation ofan inflammatory process. By contrast, apoptosis is an active process distinguished from

    necrosis by the presence of cell shrinkage, nuclear pyknosis, chromatin condensation and

    genomic fragmentation, events that occur in the absence of an inflammatory response [17].

    These two processes of neuronal death can be demonstrated following hypoxia ischemia in

    both animals and humans [2,5,17]. Since the mechanisms of neuronal necrosis versus

    apoptosis likely differ (see above), strategies to minimize brain damage in an affected infant

    following hypoxiaischemia likely will have to include interventions that target both

    processes.

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141 129

  • 7/30/2019 Pearlman 2004

    6/17

    5. Detection of neuronal injury following hypoxiaischemia with magnetic resonance

    (MR) imaging

    MR techniques have become very important in delineating evolving structural,

    metabolic and functional changes following intrapartum hypoxiaischemia [18]. Thus,

    using conventional MR, three patterns of signal abnormalities can be identified including

    injury to the thalami and/or posteriorlateral putamen with involvement of the subcortical

    white matter in the most severe cases; injury to the parasagittal gray matter and subcortical

    white matter, posterior usually more than anterior; and focal or multifocal injury [19].

    Conventional MR studies are of limited value in the early phases, i.e., hours and days

    following the insult. More recently, the use of advanced MR techniques, i.e., diffusion

    weighted imaging (DWI), has been utilized to detect early injury [20]. This technique

    measures the self-diffusion of water detected as an apparent diffusion coefficient (ADC)

    [18]. Thus, the ADC decreases with acute injury and is reflective of reduced waterdiffusivity in the tissue. Studies of adult arterial infarcts have shown that DWI signal

    changes occur within minutes of symptom onset and hours before changes become apparent

    on T1- or T2-weighted images [21]. This earlier detection of injury may facilitate

    intervention prior to irreversible injury. DWI changes may be negative if it is performed

    earlier than 24 h of life or later than 8 days of life [20]. Recently, the development of

    magnetic resonance spectroscopy (MRS) has facilitated the detection of tissue metabolism

    and in vivo biochemistry [19,20]. Thus, proton MRS (1H-MRS) can identify a spectrum of

    metabolites including lactate, creatine, N-acetylaspartate (NAA) and glutamine to name a

    few. Indeed, elevated lactate levels have been demonstrated following hypoxiaischemia in1H-MRS with regional differences, i.e., higher lactate/NAA ratios in basal ganglia than in

    occipito-parietal cortex [22]. Moreover, an early elevated lactate/creatine ratio with 1H-

    MRS coupled with a low phosphocreatine/inorganic phosphate ratio done with phosphorusMRS correlated with neurodevelopmental outcome at 1 year [23]. Thus, the data indicate

    that MRS plays an important role in the assessment of the encephalopathic infant following

    intrapartum hypoxiaischemia.

    6. Strategies to prevent ongoing injury following hypoxiaischemia

    The goals of management of a newborn infant who has sustained a hypoxicischemic

    insult and is at risk for evolving injury should include: (1) early identification of the infant

    at highest risk for evolving injury, (2) supportive care to facilitate adequate perfusion and

    nutrients to the brain and (3) consideration of interventions to ameliorate the processes ofongoing brain injury.

    Each of these approaches is briefly discussed next.

    7. Early identification of high risk infants

    The initial step in management is early identification of those infants at greatest

    risk for evolving to the syndrome of hypoxicischemic encephalopathy (HIE). This is

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141130

  • 7/30/2019 Pearlman 2004

    7/17

    a highly relevant issue because the therapeutic window, i.e., the time interval

    following hypoxiaischemia during which interventions might be efficacious in

    reducing the severity of ultimate brain injury, is likely to be short. Based onexperimental studies, it is estimated to vary from 2 to 6 h. Given this presumed short

    window of opportunity, infants must be identified as soon as possible following

    delivery in order to facilitate the implementation of early interventions. There are

    increasing data to indicate that the highest risk infant can be identified shortly after

    birth by a constellation of findings. These include evidence of a sentinel event during

    labor, i.e., fetal heart rate abnormality, a severely depressed infant (low extended

    Fig. 2. Amplitude EEG tracings depicting a normal pattern (top panel), moderate suppression (middle panel) and

    severe suppression (bottom panel).

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141 131

  • 7/30/2019 Pearlman 2004

    8/17

    Apgar score), the need for resuscitation in the delivery room (i.e., intubation, chest

    compressionFepinephrine administration), evidence of severe fetal acidemia (cord

    umbilical artery pHb7.00 and/or base deficit N

    16 mEq/l) [24], followed by evidenceof an early abnormal neurologic examination and/or abnormal assessment of cerebral

    function, i.e., integrated EEG [2527] (Fig. 2). Thus, in a prospective study of 50

    infants who had evidence of intrapartum distress, Apgar score 5 at 5 min, or cord

    arterial pH 7.00 and underwent an early neurologic examination using a modified

    Sarnat staging system (stages 2 and 3 were regarded as abnormal) and a blinded

    simultaneous a-EEG measurement, predictive values were calculated for a short-term

    abnormal outcome defined as persistent moderate to severe encephalopathy beyond 5

    days. An abnormal outcome evolved in 14 (28%) of the 50 infants. The neurologic

    examination was performed at 5F3 h after delivery. A short-term abnormal outcome

    occurred in 9 (53%) of 17 infants with initial stage 2 and in both infants with initial

    stage 3 encephalopathy. In addition, 13 infants manifested features of both stages 1and 2 encephalopathy and post hoc were classified (S12). Three of the latter infants

    (23%) developed an abnormal short-term outcome. The a-EEG was abnormal in 15

    (30%) infants, 11 (73%) of whom developed an abnormal outcome. An abnormal a-

    EEG was more specific (89% vs. 78%), had a greater positive predictive value (73%

    vs. 58%), and had similar sensitivity (79% vs. 78%) and negative predictive value

    (90% vs. 91%) when compared with an abnormal early neurologic examination. A

    combination of abnormalities had the highest specificity (94%) and positive predictive

    value (85%). Thus, a combination of the a-EEG and the neurologic examination

    shortly after birth enhances the ability to identify high-risk infants and limits the

    number of infants who would be falsely identified compared with either evaluation

    alone [27].

    8. Supportive care

    Supportive care should be directed towards the maintenance of adequate ventilation,

    avoidance of hypotension, judicious fluid management, avoidance of hypoglycemia and

    treatment of seizures. For this review, we will focus on the avoidance of hypoglycemia and

    the judicious treatment of seizures (Table 1).

    9. Control of blood glucose concentrations

    In the context of cerebral hypoxiaischemia, experimental studies suggest that both

    hyperglycemia and hypoglycemia may accentuate brain damage. Regarding hyper-

    glycemia, in adult experimental models as well as in humans, brain damage is

    accentuated. However, in immature animals subjected to cerebral hypoxiaischemia,

    hyperglycemia to blood glucose concentration of 600 mg/dl entirely prevents the

    occurrence of brain damage [28]. Regarding hypoglycemia, the effects in experimental

    animals vary and are related to the mechanisms of the hypoglycemia. Thus, insulin-

    induced hypoglycemia is detrimental to immature rat brain subjected to hypoxia

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141132

  • 7/30/2019 Pearlman 2004

    9/17

    ischemia. However, if hypoglycemia is induced by fasting, a high degree of protection is

    noted [29]. This protective effect is presumed to be secondary to the increased

    concentrations of ketone bodies, which presumably serve as alternative substrates to the

    immature brain. In a recent retrospective report, the potential contribution of initialhypoglycemia to the development of neonatal brain injury in term infants with severe

    fetal acidemia was assessed in 185 term infants admitted to neonatal intensive care with

    an umbilical arterial pHb7.00. Short-term neurologic outcome measures included death as

    a consequence of severe encephalopathy and evidence of moderate to severe

    encephalopathyFseizures. Hypoglycemia was defined as a blood glucose V40 mg/dl.

    Forty-one (22%) infants developed an abnormal neurologic outcome including 14 (34%)

    with severe HIE who died, 24 (59%) with moderate to severe HIE and 3 (7%) with

    seizures. Twenty-seven (14.5%) of the 185 infants had an initial blood sugar V40mg/dl

    (Table 2). An abnormal neurologic outcome was noted in 15/27 (56%) infants with a

    blood sugarV40 mg/dl versus 26/158 (16%) of infants with a blood sugar N40 mg/dl had

    (p=0.00003) (odds ratio (OR) 6.3 (95% CI 2.6, 15). By multivariate logistic analysis,four variables were significantly associated with abnormal outcome. However, an initial

    blood glucose V40 versus N40 mg/dl (p=0.001) (OR=18.5, 95% CI=3.1111.9) was the

    most significant variable. The other parameters, i.e., cord arterial pHV6.90 versus N6.90

    (p=0.003), OR=9.8 (CI=2.144.7), a 5-min Apgar score V5 versus N5 (p=0.006),

    OR=6.4 (CI=1.724.5) and the requirement for intubationFCPR versus neither (p=0.02),

    OR=4. 7 (CI=1.217.9) were less so [30]. These data suggest that initial hypoglycemia is

    an important risk factor for perinatal brain injury particularly in depressed term infants

    requiring resuscitation with severe fetal acidemia. Thus, in the ongoing management of

    Table 1

    A guide to the supportive management of an infant at risk for hypoxicischemic cerebral injury

    VentilationMaintain paCO2 within a normal range

    Perfusion

    Promptly treat hypotension

    Avoid hypertension

    Fluid status

    Initial fluid restriction

    Follow serum sodium and daily weights

    Blood glucose

    Maintain blood glucose within a normal range

    Avoid hypoglycemia

    Seizures

    Treat clinical seizures, particularly with an electrographic correlate

    Potential role of prophylactic phenobarbital

    Electrolyte imbalance

    Monitor serum electrolytes, calcium and magnesium

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141 133

  • 7/30/2019 Pearlman 2004

    10/17

    hypoxiaischemia, a glucose level should be screened shortly after birth and monitored

    closely. If the blood glucose is low, it should be promptly corrected.

    9.1. Seizures

    Hypoxicischemic cerebral injury is the most common cause of early onset neonatal

    seizures [2]. Although seizures are a consequence of the underlying brain injury, seizures

    activity in itself may contribute to ongoing injury. Experimental evidence strongly

    suggests that repetitive seizures disturb brain growth and development as well as increase

    the risk for subsequent epilepsy [31,32]. Despite the potential adverse effects of seizures,

    the question of which infants should be treated remains controversial [2,33]. This is a part

    related to the observation that not all clinical seizures have an electrographic correlation

    [34].

    9.2. Prophylactic phenobarbital

    Experimental data indicates that barbiturate pretreatment and even early posttreatment in

    adult animals subjected to cerebral hypoxiaischemia reduces the severity of ultimate brain

    damage. The prophylactic administration of bhigh doseQ barbiturates to infants at highest

    risk for evolving to HIE has been evaluated in two small studies with conflicting results

    [35,36]. In the first randomized study, the administration of thiopental initiated within 2 h

    and infused for 24 h did not alter the frequency of seizures, intracranial pressure or short-

    term neurodevelopmental outcome at 12 months [35]. Of importance was the observation

    that systemic hypotension occurred significantly more often in the treated group. In thesecond randomized study, phenobarbital 40 mg/kg body weight administered intravenously

    between 1 and 6 h to asphyxiated infants was associated with subsequent neuroprotection

    [36]. Although there was no difference in the frequency of seizures between the two groups

    in the neonatal period, 73% of the pre-treated infants compared to 18% of the control group

    (pb0.05) revealed normal neurodevelopmental outcome at 3-year follow-up. No adverse

    effect of phenobarbital administration was observed in this study. Thus, the role of

    prophylactic barbiturates in high-risk infants remains unclear at this current time and may

    be worthy of further investigation.

    Table 2

    Characteristics of infants with abnormal as compared to normal outcome with initial hypoglycemia

    Characteristic Abnormal outcome n=15 Normal outcome n=12 p-valueCPR 7 1 0.08

    IntubationFCPR 12 4 0.04

    5-min Apgar scoreb5 11 1 0.001

    Cord arterial pH 6.86F0.07 6.92F0.04 0.004

    Base deficit 19F4 18F2

    Initial blood pressure (mm Hg) 34F10 42F10 0.006

    Initial blood sugar (mg/dl) 18F15 17F11

    Initial temperature (8C) 36.3F0.7 36.7F0.9

    CPR=cardiopulmonary resuscitation.

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141134

  • 7/30/2019 Pearlman 2004

    11/17

    10. Potential neuroprotective strategies aimed at ameliorating secondary brain injury

    10.1. Hypothermia

    Modest systemic or selective cooling of the brain by as little as 24 8C has been shown

    to reduce the extent of tissue injury in experimental studies as well as human following

    events such as stroke, trauma or cardiac arrest [3753]. Potential mechanisms of

    neuroprotection with hypothermia include inhibition of glutamate release, reduction of

    cerebral metabolism which in turn preserves high energy phosphates, decrease in

    intracellular acidosis and lactic acid accumulation, preservation of endogenous antiox-

    idants, reduction of nitric oxide production, prevention of protein kinase inhibition,

    improvement of protein synthesis, reduction of leukotriene production, prevention of

    blood brain barrier disruption and brain edema and inhibition of apoptosis [47,49].

    Although neuroprotection following intra-ischemic hypothermia is well established, theeffects of post-ischemic hypothermia have been less certain. The latter is the typical

    scenario in the human newborn. Before initiating hypothermia as a therapeutic strategy in

    neonates, several factors had to be considered. The first relates to duration of therapy.

    Thus, brief post-ischemic hypothermia, i.e., less than 6 h results, is only partial or

    temporary neuroprotection in most experimental animals [37,49]. Prolonging post-

    ischemic hypothermia from 24 to 72 h attenuates brain damage and improves behavioral

    performance [41,42,49]. The second factor relates to the degree of hypothermia.

    Accordingly, moderate hypothermia at brain temperatures of 3234 8C initiated

    immediately or within a few hours after reperfusion and continued for 2472 h has been

    shown to favorably affect outcome in adult and newborn animals [41,42]. The third factor

    relates to the delay in initiation of therapy. Clearly, the sooner hypothermia can be

    initiated, the more likely it is to be successful. Indeed, studies indicate that, if therapy isinitiated beyond 6 h following hypoxiaischemia or following the onset of seizures, that

    hypothermia is not neuroprotective [43,44]. The fourth factor relates to the method of

    achieving hypothermia. Selective head cooling is attractive because it reduces potential

    side effects (see below) but is associated with differential gradients within the brain [54].

    Total body cooling is more likely to be associated with side effects but less temperature

    gradient within the brain. A major concern of hypothermia in newborn infants relates to

    potential adverse effects including hypoglycemia, reduction of myocardial contractility

    and ventilationperfusion mismatch, increased blood viscosity, acidbase and electrolyte

    imbalance and an increased risk of infection [49,55].

    To address the practicality and safety of mild or minimal hypothermia, pilot studies

    were conducted utilizing both selective and systemic methods to cool the brain in termneonates with moderate to severe encephalopathy [5557]. Subsequently, two large

    randomized multicenter studies have been undertaken and completed in term infants at

    highest risk for evolving to moderate and/or severe encephalopathy, using either

    selective or systemic modest hypothermia. One of these studies utilizing selective head

    cooling has reported follow up results at 18 months (Alistair Gunn-personal

    communication). No effect of hypothermia with regard to the development of cerebral

    palsy and/or death was observed for all infants treated. However, for infants who

    presented with moderate encephalopathy and without electrographic seizures, a

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141 135

  • 7/30/2019 Pearlman 2004

    12/17

    significant benefit of hypothermia was noted. No effect was observed in those infants

    with severe encephalopathy and/or early seizures. Several critical questions need to be

    addressed as a consequence of these preliminary data: (1) What is the idealtemperature for selective head cooling? (2) Which is the best mode of inducing

    hypothermia, i.e., selective head versus total body cooling? (3) How long should

    hypothermia be maintained? (4) Will a multimodal approach especially for the more

    severely affected infants be more effective than hypothermia alone?

    10.2. Oxygen free radical inhibitors and scavengers

    One therapeutic approach for the destruction of oxygen free radicals generated during

    and following hypoxiaischemia is the administration of specific enzymes known to

    degrade highly reactive radicals to a non-reactive component. Superoxide dismutase and

    catalase are antioxidant enzymes conjugated to polyethylene glycol that prolongs theircirculatory half life and facilitates penetration across the blood brain barrier [8]. Because

    of their large molecular sizes, they are restricted to the vascular space. Thus, the positive

    effects of these conjugated compounds are presumably from within cerebral vasculature by

    improving cerebral blood flow [8]. In newborn animals, neuroprotection has only been

    shown when these agents have been administered several hours prior to the hypoxic

    ischemic insult [3,58].

    A second group of free radical inhibitors that have been shown to be effective in

    experimental animals are agents that inhibit specific reactions in the production of

    xanthines. Thus, both allopurinol and oxypurinol, xanthine oxidase inhibitors, protected

    the immature rat from hypoxicischemic brain damage when the drugs were administered

    early during the recovery phase following resuscitation [3]. In a recent clinical study,

    allopurinol administered to asphyxiated infants reduced blood concentrations of oxygenfree radicals when compared to control infants [59].

    A third group of free radical inhibitors has targeted the formation of free radicals

    specifically hydroxyl radical from free iron during reperfusion [8]. Thus, desferoxamine, a

    chelating agent, prevents the formation of free radical from iron, reduces the severity of

    brain injury and improves cerebral metabolism in animal models of hypoxiaischemia

    when given during reperfusion [8,60].

    Finally, lazeroids which are nonglucocorticoid, 21-aminosteroids, preventing iron-

    dependent lipid peroxidation by scavenging peroxyl radicals have been shown to be

    neuroprotective in both adult and immature models of hypoxicischemic brain damage [3].

    10.3. Excitatory amino acid antagonists

    Given the important role of excessive stimulation of neuronal surface receptors by

    glutamate in promoting a cascade of events leading to cellular death [2,5], it has been

    logical to identify pharmacological agents that would either inhibit glutamate release or

    block its postsynaptic action.

    Glutamate receptor antagonists (i.e., NMDA, AMPA subtypes) have been extensively

    investigated in experimental animals. Non-competitive antagonists provided a reduction in

    brain damage in adult animals even when administered up to 24 h after the insult. The

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141136

  • 7/30/2019 Pearlman 2004

    13/17

    available NMDA antagonists include dizocilpine (MK-801), magnesium, phencyclidine

    (PCP), dextrometorphan and ketamine [3].

    MK-801 has been shown to be an efficacious agent in reducing the extent of hypoxicischemic brain damage in both adult and neonatal animal models [3]. This neuroprotective

    effect appears to be greater in immature compared to the adult model. This age-related

    neuroprotective effect may reflect the alterations of density and distribution of glutamate

    receptor subtypes during development of the brain.

    Magnesium is an NMDA antagonist blocking neuronal influx of Ca+ within the ion

    channel. The effect of magnesium also appears to be dependent on the maturation of the

    glutamate receptor system. Thus, in a developmental study in mice, excitotoxic neuronal

    death was limited by magnesium which was effective only after development of several

    aspects of the excitotoxic cascade, including the coupling of the calcium influx follo wing

    NMDA over stimulation, and the presence of magnesium-dependent calcium channels [61].

    The potential neuroprotective effect of magnesium has revealed conflicting data inexperimental neonatal studies. Thus, when administered shortly following an intra-

    cerebral injection of NMDA, magnesium sulfate reduced brain injury in newborn rat.

    However, other neonatal studies have failed to demonstrate neuroprotection. In a

    piglet model of asphyxia, magnesium sulfate administered 1 h following resuscitation

    failed to decrease the severity of delayed energy failure. In addition, in a near-term

    fetal lamb model, magnesium sulfate administered before and during umbilical cord

    occlusion did not influence the electrophysiologic responses or neuronal loss

    [3,62,63].

    Magnesium sulfate is an attractive agent because of its frequent use in the United State.

    It is often administered to mothers as a tocolytic agent or to prevent seizures in mothers

    with pregnancy-induced hypertension. Preliminary retrospective observation in premature

    infants noted a reduced incidence of CP at 3 years in those exposed to antenatalmagnesium sulfate [64]. A small randomized clinical study showed some subtle benefits,

    i.e., on CT imaging and improved feeding at 14 days from magnesium infusion [65].

    Clearly, future research is necessary in order to determine the potential neuroprotective

    role of magnesium.

    10.4. Prevention of nitric oxide formation

    The inhibition of NOS is another potential treatment modality to decrease brain injury.

    The experimental effects of NOS inhibitors vary and are animal species specific and

    influenced by the pharmacological agent used, the dose and timing of therapy. Thus, in

    adult experiments, the severity of neuronal loss can be reduced by the prior administrationof inhibitors of NOS activity. A similar protective effect has been observed in the

    immature rat, whereas in fetal sheep neuronal injury appears to be accentuated following

    hypoxiaischemia [3]. Further studies are clearly necessary.

    10.5. Calcium channel blocks

    The elevation of cytosolic calcium during hypoxicischemic injury makes the reduction

    of intracellular accumulation a highly desirable therapy. One strategy of preventing

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141 137

  • 7/30/2019 Pearlman 2004

    14/17

    calcium toxicity is to inhibit Ca+ influx into neurons by using calcium channel blocks

    (e.g., flunarizine, nimodipine, nicardipine). Flunarizine has been studied in fetus and

    newborn animals and the neuroprotective effect has only been demonstrated when givenprophylactically but not following hypoxicischemic insults [3]. Nicardipine was tested in

    four severely asphyxiated infants but the positive effect was counteracted by the adverse

    effects of significant hemodynamic disturbance [66]. Indeed, current calcium channel

    blocks are in general contraindicated in the neonate and young infant in the United States

    because of significant adverse cardiovascular effects.

    10.6. Other studied therapies in immature animals

    Other avenues of potential neuroprotection include platelet-activating factor antagonists

    [67], adenosinergic agents [68], monosialoganglioside GM1 [69], growth factors, e.g.,

    nerve growth factor (NGF) [70], insulin growth factor-1 [71] and blocking the apoptoticpathways, i.e., minocycline [72], erythropoeitin, etc. [73,74].

    11. Conclusions

    Much progress has been made toward understanding the mechanisms contributing to

    ongoing brain injury following hypoxiaischemia. This should facilitate more specific

    pharmacologic intervention strategies that might provide neuroprotection during the

    reperfusion phase of injury. Early identification of infants at highest risk for brain injury

    is critical if such interventions are to be successful. Moreover, given the small number of

    asphyxiated full-term infants that are likely to be admitted to any one neonatal intensive

    care unit over an extended time period implores a continued multicenter treatmentapproach to allow inclusion of an adequate number of infants to achieve statistical

    validity.

    References

    [1] Perlman JM. Intrapartum hypoxicischemic cerebral injury and subsequent cerebral palsy: medico-legal

    issues. Pediatrics 1997;99:851 9.

    [2] Volpe JJ. Hypoxicischemic encephalopathy. In: Volpe JJ, editor. Neurology of the newborn. Philadelphia

    (PA)7 WB Saunders; 2001.

    [3] Vannucci RC, Perlman JM. Interventions for perinatal hypoxicischemic encephalopathy. Pediatrics

    1997;100:100414.

    [4] Wyatt JS, Edwards AD, Azzopardi D, Reynolds EOR. Magnetic resonance and near infrared spectroscopy

    for investigation of perinatal hypoxicischemic brain injury. Arch Dis Child 1989;64:953 63.

    [5] Grow J, Barks DE. Pathogenesis of hypoxicischemic cerebral injury in the term infant: current concepts.

    Clin Perinatol 2002;29:585 602.

    [6] Lorek A, Takei Y, Cady EB, Wyatt JS, Penrice J, Edwards AD, et al. Delayed (bsecondaryQ) cerebral energy

    failure after acute hypoxiaischemia in the newborn piglet: continuous 48-hour studies by phosphorus

    magnetic resonance spectroscopy. Pediatr Res 1994;36:699 706.

    [7] Roth SC, Baudin J, Cady E, Johal K, Townsend JP, Wyatt JS, et al. Relation of deranged neonatal cerebral

    oxidative metabolism with neurodevelopmental outcome and head circumference at four years. Dev Med

    Child Neurol 1997;39:71825.

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141138

  • 7/30/2019 Pearlman 2004

    15/17

    [8] Palmer C. Hypoxicischemic encephalopathy. Therapeutic approaches against microvascular injury, and

    role of neutrophils, PAF, and free radicals. Clin Perinatol 1995;22:481517.

    [9] Hagberg H, Gilliaud E, Bona E. Enhanced expression of IL1 and IL6 m-RNA and bioactive proteins after

    hypoxia ischemia in neonatal rats. Pediatr Research 1996;40:6039.

    [10] Bona E, Anderson AL. Chemokines and inflammatory cell response to hypoxia ischemia in immature rats.

    Pediatr Research 1999;45:5009.

    [11] Eklind S, Mallard C, Leverin AL. Bacterial endotoxin sensitizes the immature brain to hypoxic ischemic

    injury. Europ J Neuroscience 2001;13(6):1101 6.

    [12] Hagan P, Barks GDE, Yabut M, et al. Adenovirus mediated overexpression of IL1 receptor antagonist

    reduces susceptibility to excitotoxic brain injury in perinatal rats. Neuroscience 1996;75:1033 45.

    [13] Yanasaki Y, Shoozurhara H, Onodera H, et al. Blocking of IL-1 activity is a beneficial approach to ischemia

    and brain edema formation. Acta Neurochir 1995;60:3002.

    [14] Bruce AJ, Boling W, Kindy S. Altered neuronal and microglial responses to excitotoxic and ischemic brain

    injury in mice lacking TNF receptors. Nature Med 1996;2:78894.

    [15] Loddick SA, Turnbill AV, Rothwell NJ. Cerebral interleukin 1 is neuroprotective against permanent focal

    ischemia in the rat. J Cerebral Blood Flow and Metabolism 1998;18:1769.

    [16] Bonfold E, Krainc D, Ankarcrona M, Nicotera P, Lipton SA. Apoptosis and necrosis: two distinct events,induced respectively by mild and intense insults with N methyl-dospartate or nitric oxide/superoxide in

    cortical cell cultures. Proc Natl Acad Sci U S A 1995;92:71626.

    [17] Edwards AD, Mehmet H. Apoptosis in perinatal hypoxicischemic cerebral damage. Neuropathol Appl

    Neurobiol 1996;22:494 8.

    [18] Huppi PS. Advances in postnatal neuroimaging:relevance to pathogenesis and treatment of brain injury. Clin

    Perinatol 2002;29:827 56.

    [19] Ment LR, Bada HS, Barnes P, et al. Practice parameter: neuroimaging of the neonate: report of the quality

    standards subcommittee of the American Academy of Neurology and the Practice Committee of the Child

    Neurology Society. Neurology 2002;58:1726 38.

    [20] Cowan FM, Pennock JM, Hanrahan JD, et al. Early detection of cerebral infarction and hypoxic ischemic

    encephalopathy in neonates using diffusion-weighted magnetic resonance imaging. Neuropediatrics

    1994;25:1725.

    [21] Albers GW, Lansberg MG, Norbash AM, et al. Yield of diffusion-weighted MRI for detection of potentially

    relevant findings in stroke patients. Neurology 2000;54:15627.[22] Groenendaal F, Veenhoven RH, van der Grond J, et al. Cerebral lactate and N-acetyl-aspartate/choline ratios

    in asphyxiated full-term neonates demonstrated in vivo using proton magnetic resonance spectroscopy.

    Pediatr Res 1994;34:14851.

    [23] Hanrahan JD, Cox IJ, Edwards AD. Persistent increases in cerebral lactate concentration after birth

    asphyxia. Pediatr Res 1998;44:308 11.

    [24] Risser JM, Perlman R. Can asphyxiated infants at risk for neonatal seizures be rapidly identified by current

    high-risk markers? Pediatrics 1996;97:456 62.

    [25] Hellstrom-Westas L, Rosen I, Svenningsen NW. Predictive value of early continuous amplitude

    integrated EEG recordings on outcome after severe birth asphyxia in full term infants. Arch Dis Child

    1995;72:F348.

    [26] al Naqeeb N, Edwards AD, Cowan FM, Azzopardi D. Assessment of neonatal encephalopathy by

    amplitude-integrated electroencephalography. Pediatrics 1999;103:12639.

    [27] Shalak L, Corbett R, Laptook AR, Perlman JM. Amplitude-integrated electroencephalography coupled with

    an early neurologic examination enhances prediction of term infants at risk for persistent encephalopathy.

    Pediatrics 2003;111:351 7.

    [28] Vannucci RC, Mujsce DJ. Effect of glucose on perinatal hypoxicischemic brain damage. Biol Neonate

    1992;62:21524.

    [29] Yager JY. Hypoglycemic injury to the immature brain. Clin Perinatol 2002;29:651 74.

    [30] Salhab W, Wyckoff M, Laptook AR, Perlman JM. Initial hypoglycemia and neonatal brain injury in term

    infants with severe fetal acidemia. Pediatrics 2004;114:361 6.

    [31] Dzhala V, Ben-Ari Y, Khazipov R. Seizures accelerate anoxia-induced neuronal death in the neonatal rat

    hippocampus. Ann Neurol 2000;48:63240.

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141 139

  • 7/30/2019 Pearlman 2004

    16/17

    [32] Holmes GL, Gairsa JL, Chevassus AL, Ben-Ari Y. Consequences of neonatal seizures in the rat:

    morphological and behavioral effects. Ann Neurol 1998;44:845 57.

    [33] Mizrahi EM. Consensus and controversy in the clinical management of neonatal seizures. Clin Perinatol

    1989;16:485500.

    [34] Scher MS, Aso K, Beggerly ME, et al. Electrographic seizures in preterm and fullterm infants neonates:

    clinical correlates associated brain lesions and risk for neurologic sequelae. Pediatrics 1993;91:128 34.

    [35] Goldberg RN, Moscoso P, Bauer CR, Bloom FL, Curless RG, Burke B, et al. Use of barbiturate therapy in

    severe perinatal asphyxia: a randomized controlled trial. J Pediatr 1986;109:8516.

    [36] Hall RT, Hall FK, Daily DK. High-dose phenobarbital therapy in term newborn infants with severe perinatal

    asphyxia: a randomized prospective study with three-year follow-up. J Pediatr 1998;132:3458.

    [37] Minamisawa H, Smith ML, Siesjo BK. The effects of mild hyperthermia and hypothermia on brain damage

    following 5, 10, and 15 minutes of forebrain ischemia. Ann Neurol 1990;28:2633.

    [38] Yager J, Towfighi J, Vannucci RC. Influence of mild hypothermia on hypoxicischemic brain damage in the

    immature rat. Pediatr Res 1993;35:5259.

    [39] Laptook AR, Corbett RJT, Sterett R, et al., Modest hypothermia provides partial neuroprotection for

    ischemice neonatal brain. Pediatr Res 35:43642.

    [40] Thorensen M, Bagenholm R, Loberg EM, et al. Posthypoxic cooling of neonatal rats provides protectionagainst brain injury. Arch Dis Child 1996;74:F3.

    [41] Gunn TR, Gunn AJ, de Haan Tannia R, Harmen H, et al. Dramatic neuronal rescue with prolonged selective

    head cooling after ischemia in fetal lambs. J Clin Invest 1997;99:248 56.

    [42] Gunn TR, Gunn AJ, Gunning TR, Mark I. Neuroprotection with prolonged head cooling started before

    postischemic seizures in fetal sheep. Pediatrics 1998;102:1098 106.

    [43] Gunn AJ, Bennet L, et al. Cerebral hypothermia is not neuroprotective when started after postischemic

    seizures in fetal sheep. Pediatr Res 1999;46(3):27480.

    [44] Gunn AJ. Cerebral hypothermia for prevention of brain injury following perinatal asphyxia. Curr Opin

    Pediatr 2000;12(2):111 5.

    [45] Colbourne F, Sutherland G, Corbett D. Postischemic hypothermia: a critical appraisal with implications for

    clinical treatment. Mol Neurobiol 1997;14:171 201.

    [46] Edwards AD, Wyatt JS, Thoresen M. Treatment of hypoxicischaemic brain damage by moderate

    hypothermia. Arch Dis Child Fetal Neonatal Ed 1998;78:F8591.

    [47] Laptook AR, Corbett RJT. Therapeutic hypothermia: a potential neuroprotective and resuscitative strategyfor neonatal hypoxiaischemia. Prenat Neonat Med 1996;1:199212.

    [48] Towfighi J, Housman C, Heitjan DF, Vannucci RC, Yager JY. The effect of focal cerebral cooling on

    perinatal hypoxicischemic brain damage. Acta Neuropathol 1994;87:598 604.

    [49] Laptook AR, Corbett RJT. The effects of temperature on hypoxicischemic brain injury. Clin Perinat

    2002;29:6239.

    [50] Marion DW, Penrod LE, Kelsey SF, Obrist WD, Kochanek PM, Palmer AM, et al. Treatment of traumatic

    brain injury with moderate hypothermia. N Engl J Med 1997;336:540 6.

    [51] Bernard SAT. Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia.

    NEJM 2002;346:557 63.

    [52] Hypothermia Group. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest.

    NEJM 2002;346:549 56.

    [53] Safar PJ, Therapeutic Hypothermia after Cardiac Arrest. NEJM 346;6123.

    [54] Laptook AR, Shalak L, Corbett RJT. Differences in brain temperature and cerebral blood flow during

    selective head versus whole-body cooling. Pediatrics 2001;108:1103 10.

    [55] Thoresen Mariane, Whitelaw Andrew. Cardiovascular changes during mild therapeutic hypothermia and

    rewarming in infants with hypoxicischemic encephalopathy. Pediatrics 2000;106:92 9.

    [56] Gunn AJ, Gluckman PD, Gunn TR. Selective head cooling in newborn infants after perinatal asphyxia. A

    safety study. Pediatrics 1998;102:885 92.

    [57] Azzopardi D, Robertson NJ, Cowan FM, et al. Pilot study of treatment with whole body hypothermia for

    neonatal encephalopathy. Pediatrics 2000;106:68494.

    [58] Shimizu K, Rajapakse N, Horiguchi T. Neuroprotection against hypoxiaischemia in neonatal rat brain by

    novel superoxide dismutase mimetics. Neuroscience Letters 2003;346:41 4.

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141140

  • 7/30/2019 Pearlman 2004

    17/17

    [59] Van Bel F, Shadid M, Moison RM, Dorrepaal CA, Fontijn J, Monteiro L, et al. Effect of allopurinol on post-

    asphyxial free radical formation, cerebral hemodynamics and electrical brain activity. Pediatrics

    1998;101:18593.

    [60] Peeters-Schulte C, et al. Effects of allopurinol and desferoxamine on reperfusion injury of the brain in

    newborn piglets after neonatal hypoxiaischemia. Pediatr Res 2003;54:516 22.

    [61] Marret S, Gressens P, Gadisseux JF, Evrard P. Prevention by magnesium of excitotoxic neuronal death in the

    developing brain: an animal model of clinical intervention studies. Dev Med Child Neurol 1995;37:47384.

    [62] Penrice J, Amess PN, Punwani S, Wylezinska M, Tyszczuk L, DSouza P, et al. Magnesium sulfate after

    transient hypoxiaischemia fails to prevent delayed cerebral energy failure in the newborn piglet. Pediatr

    Res 1997;41:4437.

    [63] de Haan HH, Gunn AJ, Williams CE, Heymann MA, Gluckman PD. Magnesium sulfate therapy during

    asphyxia in near-term fetal lambs does not compromise the fetus but does not reduce cerebral injury. Am J

    Obstet Gynecol 1997;176:1827.

    [64] Nelson KB, Grether JK. Can magnesium sulfate reduce the risk of cerebral palsy in very low birthweight

    infants? Pediatrics 1995;95:263 9.

    [65] Ischiba H. Randomized controlled trial of magnesium sulfate infusion for severe birth asphyxia. Pediatr Int

    2002;44:5059.[66] Levene MI, Gibson NA, Fenton AC, Papathoma E, Barnett D. The use of a calcium-channel blocker,

    nicardipine, for severely asphyxiated newborn infants. Dev Med Child Neurol 1990;32:567 74.

    [67] Liu XH, Eun BL, Silverstein FS, Barks JD. The platelet-activating factor antagonist BN 52021 attenuates

    hypoxicischemic brain injury in the immature rat. Pediatr Res 1996;40:797803.

    [68] Halle JN, Kasper CE, Gidday JM, Koos BJ. Enhancing adenosine A1 receptor binding reduces hypoxic

    ischemic brain injury in newborn rats. Brain Res 1997;759:309 12.

    [69] Tan WK, Williams CE, Mallard CE, Gluckman PD. Monosialoganglioside GM1 treatment after a hypoxic

    ischemic episode reduces the vulnerability of the fetal sheep brain to subsequent injuries. Am J Obstet

    Gynecol 1994;170:6639.

    [70] Holtzman DM, Sheldon RA, Jaffe W, Cheng Y, Ferriero DM. Nerve growth factor protects the neonatal

    brain against hypoxicischemic injury. Ann Neurol 1996;39:114 22.

    [71] Johnston BM, Mallard EC, Williams CE, Gluckman PD. Insulin-like growth factor-1 is a potent neuronal

    rescue agent after hypoxicischemic injury in fetal lambs. J Clin Invest 1996;97:300 8.

    [72] Arvin K, et al. Minocycline markedly protects the neonatal brain against hypoxicischemic injury. AnnNeurol 2002;52:54 61.

    [73] Aydin A, et al. Erythropoietin exerts neuroprotective effect in neonatal rat model of hypoxicischemic brain

    injury. Brain and Development 2003;25:4948.

    [74] Kumral A, et al, Neuroprotective effect of erythropoietin on hypoxicischemic brain injury in neonatal rats

    Biology of the Neonate 2203;83:2248.

    L. Shalak, J.M. Perlman / Early Human Development 80 (2004) 125141 141