6
Best Practice Guideline article Obstetric aspects of hypoxic ischemic encephalopathy Sailesh Kumar 1 , Sara Paterson-Brown Queen Charlotte's and Chelsea Hospital, Imperial College London, Du Cane Road, London W12 0HS, United Kingdom abstract article info Keywords: Hypoxic ischemic encephalopathy Neonatal encephalopathy Intrapartum asphyxia Cerebral palsy Fetal heart monitoring Hypoxic ischemic encephalopathy (HIE) describes neonatal encephalopathy that is caused by intrapartum asphyxia and it can result in the long term sequelae of cerebral palsy which is a major cause of disability. The incidence of cerebral palsy has not changed over the last few decades and the challenge to obstetricians remains how best to recognise those babies at risk of this intrapartum insult both before and during labour. Many associations and risk factors are unavoidable or unrecognisable, and others are fairly common and associated with poor predictive value. Intrapartum fetal heart monitoring remains the main focus of attention but how this is best achieved is still the subject of research. Computerised decision support systems built into fetal heart rate monitoring and non-invasive fetal ECG signal pick-up are currently being explored. © 2010 Elsevier Ireland Ltd. All rights reserved. Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 339 2. Diagnosis of hypoxic ischemic encephalopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 340 3. Patterns of brain injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 340 4. Risk factors for perinatal hypoxia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341 5. Prevention of hypoxic ischemic encephalopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341 5.1. Antenatal detection of fetal growth restriction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341 5.2. Intrapartum care . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341 5.2.1. Minimising intrauterine infection/inammation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341 5.2.2. Fetal heart rate monitoring . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342 5.2.3. More liberal use of caesarean section . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342 5.2.4. Staff training, intrapartum guidelines and clinical governance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342 6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343 1. Introduction Hypoxic ischemic encephalopathy (HIE) and neonatal encepha- lopathy and their long term sequelae of cerebral palsy are major causes of disability in term and near term infants. They feature prominently in medico-legal claims against the health service both in terms of their quantum but also for the length of time taken to resolve. Their prevalence is estimated at 16 per 1000 live full-term births [1]. Neonatal encephalopathy is a clinical syndrome of disturbed neu- rologic function of the term and near term infant in the early neonatal period, manifested by respiratory difculties, depression of tone and reexes, obtundation, and frequently by seizures [2]. It is classied as mild, moderate or severe (or stage 1, 2 or 3) according to the criteria by Sarnat and Sarnat (1976) [3] and is based on the baby's response to handling, consciousness level, abnormalities of tone or reexes, pres- ence of seizures and the duration of the symptoms after birth. The etiology of neonatal encephalopathy is varied, with many genetic and metabolic conditions presenting with similar clinical signs [4]. The disorder is only called hypoxic ischemic encephalopathy if there is evidence that intrapartum asphyxia is the cause of the encephalop- athy resulting in neurologic depression or seizures. However, there is often confusion between the two conditions as they are frequently used interchangeably in the literature. Although it was previously believed that intrapartum asphyxia was the main causative factor for the development of cerebral palsy, Early Human Development 86 (2010) 339344 Corresponding author. Tel.: + 44 208 3833998; fax: + 44 208 3833507. E-mail addresses: [email protected] (S. Kumar), [email protected] (S. Paterson-Brown). 1 Tel.: +44 208 3833998; fax: +44 208 3833507. 0378-3782/$ see front matter © 2010 Elsevier Ireland Ltd. All rights reserved. doi:10.1016/j.earlhumdev.2010.05.009 Contents lists available at ScienceDirect Early Human Development journal homepage: www.elsevier.com/locate/earlhumdev

Obstetric aspects of hypoxic ischemic encephalopathy

Embed Size (px)

Citation preview

Early Human Development 86 (2010) 339–344

Contents lists available at ScienceDirect

Early Human Development

j ourna l homepage: www.e lsev ie r.com/ locate /ear lhumdev

Best Practice Guideline article

Obstetric aspects of hypoxic ischemic encephalopathy

Sailesh Kumar 1, Sara Paterson-Brown ⁎Queen Charlotte's and Chelsea Hospital, Imperial College London, Du Cane Road, London W12 0HS, United Kingdom

⁎ Corresponding author. Tel.: +44 208 3833998; fax:E-mail addresses: [email protected] (S. K

[email protected] (S. Paterson-Brow1 Tel.: +44 208 3833998; fax: +44 208 3833507.

0378-3782/$ – see front matter © 2010 Elsevier Irelanddoi:10.1016/j.earlhumdev.2010.05.009

a b s t r a c t

a r t i c l e i n f o

Keywords:

Hypoxic ischemic encephalopathyNeonatal encephalopathyIntrapartum asphyxiaCerebral palsyFetal heart monitoring

Hypoxic ischemic encephalopathy (HIE) describes neonatal encephalopathy that is caused by intrapartumasphyxia and it can result in the long term sequelae of cerebral palsy which is a major cause of disability. Theincidence of cerebral palsy has not changed over the last few decades and the challenge to obstetriciansremains how best to recognise those babies at risk of this intrapartum insult both before and during labour.Many associations and risk factors are unavoidable or unrecognisable, and others are fairly common andassociated with poor predictive value. Intrapartum fetal heart monitoring remains the main focus ofattention but how this is best achieved is still the subject of research. Computerised decision support systemsbuilt into fetal heart rate monitoring and non-invasive fetal ECG signal pick-up are currently being explored.

+44 208 3833507.umar),n).

Ltd. All rights reserved.

© 2010 Elsevier Ireland Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3392. Diagnosis of hypoxic ischemic encephalopathy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3403. Patterns of brain injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3404. Risk factors for perinatal hypoxia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3415. Prevention of hypoxic ischemic encephalopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341

5.1. Antenatal detection of fetal growth restriction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3415.2. Intrapartum care . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341

5.2.1. Minimising intrauterine infection/inflammation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3415.2.2. Fetal heart rate monitoring . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3425.2.3. More liberal use of caesarean section . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3425.2.4. Staff training, intrapartum guidelines and clinical governance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342

6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343

1. Introduction

Hypoxic ischemic encephalopathy (HIE) and neonatal encepha-lopathy and their long term sequelae of cerebral palsy aremajor causesof disability in term and near term infants. They feature prominentlyin medico-legal claims against the health service both in terms oftheir quantum but also for the length of time taken to resolve. Theirprevalence is estimated at 1–6 per 1000 live full-term births [1].

Neonatal encephalopathy is a clinical syndrome of disturbed neu-rologic function of the term and near term infant in the early neonatal

period, manifested by respiratory difficulties, depression of tone andreflexes, obtundation, and frequently by seizures [2]. It is classified asmild, moderate or severe (or stage 1, 2 or 3) according to the criteriaby Sarnat and Sarnat (1976) [3] and is based on the baby's response tohandling, consciousness level, abnormalities of tone or reflexes, pres-ence of seizures and the duration of the symptoms after birth. Theetiology of neonatal encephalopathy is varied, with many genetic andmetabolic conditions presenting with similar clinical signs [4]. Thedisorder is only called hypoxic ischemic encephalopathy if there isevidence that intrapartum asphyxia is the cause of the encephalop-athy resulting in neurologic depression or seizures. However, thereis often confusion between the two conditions as they are frequentlyused interchangeably in the literature.

Although it was previously believed that intrapartum asphyxiawas the main causative factor for the development of cerebral palsy,

340 S. Kumar, S. Paterson-Brown / Early Human Development 86 (2010) 339–344

more recentpopulationbased studies have shown intrapartumhypoxia/ischemia to be present in only 8–28%of cases [5,6]. Themajority (65%)ofchildrenwith cerebral palsy arebornat term [7]. Despite thewidespreaduse of electronic fetal monitoring and increased caesarean section ratesthe incidence of cerebral palsy has not decreased in developed countriesover the past 30 years [8].

2. Diagnosis of hypoxic ischemic encephalopathy

There have been three consensus statements from various bodiesaddressing the diagnosis of intrapartum asphyxia [9–11] (Table 1).These consensus statements emphasise the use of multiple markersfor the diagnosis, with both essential criteria and supportive evidencerequired before the diagnosis can be made. Some markers are used todiagnose the presence of asphyxia and some the timing of asphyxia.

When intrapartum asphyxia was defined as an umbilical arterialpHb7.0 the incidence at termwas3.7per 1000 live births. The incidenceof HIE at term was 2.5 per 1000 live births and the proportion of casesof cerebral palsy associated with intrapartum asphyxia was 14.5% (seesystematic review by Graham et al. 2008) [12].

The most objective assessment of the presence of intrapartumhypoxia–ischemia is metabolic acidosis in umbilical arterial blood atthe time of delivery [9–11]. Term infants have a mean (SD) umbilicalarterial pH of 7.27±0.07 and base excess of −2.7±2.8 mM [13]. Innormal labour, umbilical arterial pH decreases and the base deficitincreases. Although respiratory acidosis can be quickly corrected bythe neonate, the fetus is unable to tolerate metabolic acidosis for aprolonged period of time. An increased risk of neurologic morbidityis usually seen when the umbilical arterial pH is b7.0 [14] or if thebase deficit is N12 mM [15]. Furthermore, neonatal seizures, otherneurologic abnormalities, and death are significantly more commonin neonates with a pHb7.0 [14,16]. Paradoxically, in some studies,more than two-thirds of newborn infants with an umbilical arterypHb7.0 were classified as normal on discharge from the hospital[14,17]. Of neonates with this degree of acidosis, 23.1% had neonatalneurologic morbidity or mortality [12].

Defining intrapartum hypoxia–ischemia as a cord pHb7.0 is notuniversally agreed on, and some investigators have reported cases ofintrapartum hypoxic ischemic injury occurring in neonates with cordpHN7.0 [12]. Indeed it can be argued that there is no single umbilicalarterial pH value that clearly distinguishes between cases that haveintrapartum hypoxia/ischemia and those that do not. Nevertheless, itis generally agreed that the risk for neonatal complications increasesas the pH at birth decreases.

Other parameters used to diagnose intrapartum hypoxia/HIE in-clude a low Apgar score, the presence of meconium stained liquor,fetal heart rate abnormalities from electronic monitoring or the needfor immediate neonatal resuscitation. These parameters are not spe-cific or sensitive enough for the diagnosis of either fetal hypoxia or

Table 1Consensus statements for the diagnosis of intrapartum hypoxia as the etiology for cerebral

American Academy of Paediatrics/American Collegeof Obstetrics and Gynaecology (1996) [10]

International Cerebral Palsy Task F

Essential criteria Essential criteria• Profound metabolic acidosis (pHb7.0) in anumbilical artery sample

• Metabolic acidosis in early neonaand base deficit N 12 mmol/L)

• Apgar scoreb3 after 5 min • Moderate or severe encephalopat• Neonatal encephalopathy • Cerebral palsy of spastic quadripl• Multi-organ system dysfunction

Criteria suggestive of intrapartum tim• Sentinel event• Abrupt change in fetal heart rate• Apgar score b6 beyond 5 min• Multi-system involvement• Imaging evidence

acidemia as they can be influenced by many other factors. For ex-ample, the Apgar score can be low because of intrapartum maternalsedation or anaesthesia, congenital malformations (e.g. neuromus-cular disorders), objectivity of the individual assigning the score,resuscitative efforts, and the presence of infection [12].

Although objective grading systems now exist for neonatalencephalopathy, older studies have used numerous definitions forintrapartum hypoxia/ischemia, which have contributed to inconsis-tencies in outcome and proposed etiology. Regardless of the specificdefinition used, the majority of cases of cerebral palsy are not asso-ciated with intrapartum hypoxia/ischemia.

3. Patterns of brain injury

Despite a similar etiology, the preterm and term brains respondvery differently to hypoxia/ischemia. Although these differences arenot absolute, the preterm brain is more susceptible to white matterabnormalities while the term brain exhibits primarily grey matterinjury. These differences cause the development of periventricularleukomalacia (PVL) in the preterm infant and that of hypoxic enceph-alopathy, seizures, and stroke in the term infant. The gestation ofgreatest susceptibility to PVL is between 24 and 32 weeks when thewhite matter is predominantly populated by oligodendrocyte pre-cursors and immature oligodendrocytes (preoligodendrocytes) [18].This lesion occurs almost exclusively in neonates b34 weeks, and 60–100% of survivors develop cerebral palsy [19]. PVL represents thetypical response of the preterm brain to an insult, either a hypoxic/ischemic injury (from reduced cerebral blood flow) or one caused bydamage from cytokines associated with infection. In preterm infantsthe cerebral white matter is particularly vulnerable because of itsdecreased blood flow (it receives only 25% of the blood flow of corticalgray matter), its inability to autoregulate perfusion, and a decreasednumber of anastomoses between the short and long penetrating ar-teries that provide its blood supply. The vascular end zones in thewhite matter are the sites for focal necroses of periventricularleukomalacia and white matter atrophy producing ventriculomegaly.MRI features include a reduced bulk of white matter with scallopedventricular margins and gliosis in the periventricular region. Preterminfants develop spastic diplegic cerebral palsy as a consequence ofthese cerebral injuries.

Term infants can develop athetoid (dyskinetic) cerebral palsy orspastic quadriplegic cerebral palsy as a consequence of intrapartumhypoxia. In the term or near term brain, a short period of profoundhypoxic ischemia causes injury to parts of the brain with a highmetabolic rate, high blood supply, high glucose metabolism and alarge number of excitatory glutaminergic neurons [20,21]. Theseregions include the posterior putamina of the lentiform nuclei, theventro-lateral nuclei of the thalami, the hippocampus and the peri-Rolandic cortex [22,23]. The precise pattern and extent of injury

palsy [9–11].

orce (1999) [9] American College of Obstetrics and Gynaecology(2003) [11]

Essential criteriatal blood sample (pHb7.0 •Metabolic acidosis (pHb7.0 andbasedeficitN12 mmol/L)

in umbilical artery samplehy • Moderate or severe encephalopathyegia or dyskinetic type • Cerebral palsy of spastic quadriplegia or dyskinetic type

• Exclusion of other pathologies of cerebral palsying

Criteria suggestive of intrapartum timing• Sentinel event• Abrupt change in fetal heart rate• Apgar score b6 beyond 5 min• Multi-system failure within 72 h of life• Imaging evidence

341S. Kumar, S. Paterson-Brown / Early Human Development 86 (2010) 339–344

depends on the severity and duration of the insult. Sometimes thewhitematter of the corona radiata and the optic radiations are damaged, andon occasion, there is involvement of the hippocampus [24,25].

In spastic quadriplegic cerebral palsy, parasagittal cerebral injuryor damage to the deep greymatterwithwhitematter involvement andcerebral atrophy is seen. Some children occasionally show changesconsistent with periventricular leukomalacia (PVL). Parasagittalcerebral injury occurs because of systemic and cerebral hypotensionand hypoperfusion. A prolonged period of mild to moderate hy-potension can cause damage to the brain in the parasagittal zones(watershed areas) that are supplied by the anterior, middle andposterior cerebral arteries. This pattern of injury is characterised bynecrosis of the cortex and adjacent whitematter and involves both theanterior and parieto-occipital regions. Under perfusion of the gyrileads to shrinkage at the bases and the development of ulegyria areaction that is specific to the term or near term brain [26]. There isconsiderable overlap between these two types of termbrain injury andthe clinical features may be similarly consistent.

In addition to the above, both preterm and term infants can de-velop hemiplegic cerebral palsy. However unlike athetoid or spasticcerebral palsy, hemiplegic cerebral palsy is not considered to be dueto intrapartum asphyxia. A common cause is infarction of the regionusually supplied by the left middle cerebral artery. The incidenceof neonatal stroke in term infants is estimated at 1 in 4000 cases [27].Symptoms may be mild in some infants and not recognized, althoughfocal seizures may often be present. Babies with more extensiveinfarcts may develop encephalopathy [28,29]. Risk factors for anarterial stroke in the newborn period include a prolonged labour withan occipito-posterior position, umbilical cord occlusion and inheritedthrombophilias [27,30,31].

4. Risk factors for perinatal hypoxia

Although there has been much optimism that the incidence ofcerebral palsy was likely to decrease with improvements in obstetricand neonatal care, there has been no consistent decrease in its fre-quency in the last 30 years [8] and the incidence of dyskinetic cerebralpalsy usually caused by birth asphyxia has not declined [32]. Thisdisappointment may be partially explained by the multifactorial riskfactors associated with this condition and the fact that many areunavoidable and others difficult to detect or treat (Table 2).

One of the aims of antenatal care is to screen women early in theirpregnancy to detect risk factors for adverse outcomes and then tominimise their effects with appropriate monitoring and intervention.Examples of such an approach include: Blood Pressure (BP) measure-ment and urinalysis to detect pre-eclampsia with subsequent blood

Table 2Risk factors for neonatal encephalopathy [4,25,33].

Pre-pregnancy factors Antenatalfactors

Fetal factors Intrapartum factors

Socioeconomic status Bleeding Multiplepregnancy

Infection

Family history of seizures Hypertensivedisorders

Chromosomalanomaly

Placental bleeding

Family history ofneurological disease

Abnormalplacentation

Congenitalabnormality

Feto-maternalhemorrhage

Fertility treatment Viral illness Fetal growthrestriction

Bleeding fromvasa praevia

Maternal thyroid disease Bacterialinfection

Prematurity Uterine rupture

Maternal obesity Coagulationdisorders

Coagulationdisorders

Cord accident

Postmaturity Maternal collapseBreechpresentation

Prolonged labour

Male sex Oxytocin abuse

pressure control and timely delivery; and abdominal palpation withsymphysio-fundal height measurement to detect fetal growth re-striction. While the former is reasonably straightforward the latter isvery much an inexact science (made harder by increasing maternalobesity) and therefore although severe growth restriction is a strongrisk factor for neonatal encephalopathy [33] it often goes undetected.

The importance of intrapartum factors on cerebral palsy remainsuncertain. Blair and Stanley [5] considered that only 8% of all ofthe children with spastic cerebral palsy had intrapartum asphyxia as apossible cause of their brain injury. More recently Badawi et al. (1998)[33,34] showed in a case–control study from Western Australia thatthe causes of moderate or severe neonatal encephalopathy wereheterogeneous and many etiological pathways start either pre-conception or in the antepartum period (see review by Kurinczuk inthis issue). Intrapartum events may also be responsible and a recentstudy [25] found no evidence of long-standing brain damage in acohort of babies imaged after suffering a sentinel intrapartum eventsuggesting that brain injury occurred as a result of such an eventrather than from a pre-existing cause.

The wide variation in the effect of hypoxia/ischemia on thenewborn brain suggests the possibility that genetic factors may alsoplay a significant role. Animal models of HIE show wide inter-strainvariability in the severity of injury after an identical insult [35,36].Gender differences of response to HIE have also been observed [37].There is enough evidence now to support the view that a variety ofgenetic factors may interplay in the perinatal period to increase thevulnerability of the developing brain to hypoxic/ischemic injury.

5. Prevention of hypoxic ischemic encephalopathy

5.1. Antenatal detection of fetal growth restriction

Detecting risk factors in the antenatal period can enable closefollow-up of women whose babies are at risk and this in turn canallow appropriate decisions about timing, mode and place of deliveryto bemade. Improving the clinical diagnosis of fetal growth restrictionwould be a step forward in this context, but routine late pregnancygrowth scans are not cost effective [38] and routine Doppler studiesare not beneficial [39]. Furthermore, late scans in pregnancy are as-sociated with a small increase in caesarean section rates [39]. Thedetection of the small baby remains very much a clinical skill. Agrowth restricted baby at term is more at risk of intrapartum hypoxic/ischemic injury and therefore if the diagnosis has been made thethreshold for operative delivery may be lower, but many small babiesare delivered safely vaginally and there is constant pressure on ob-stetricians to avoid ‘unnecessary’ caesarean sections.

5.2. Intrapartum care

Although the majority of fetal neurological injuries predate labour,the intrapartum period is a time of risk for asphyxia and asphyxia-related morbidity and mortality. Intrapartum care aims to identifythose babies at risk of hypoxia to recognise when signs develop andthen to intervene in a timely way.

5.2.1. Minimising intrauterine infection/inflammationIt has been proposed that cytokines may be the final common

mediators of brain injury that is initiated by hypoxia/ischemia, re-perfusion, and infection [40]. Furthermore there have been manypublications linking the presence of maternal fever or placental in-flammation (chorioamnionitis)with adverse neurologic outcome [41].The fetal inflammatory response and cytokine mediated injury effecton oligodendrocytes, has been implicated in the white matter damagethat occurs in preterm infants. A similar association is also present interm infants. The effect of any intrapartumhypoxia/ischemia is greaterif maternal pyrexia is present in labour.

342 S. Kumar, S. Paterson-Brown / Early Human Development 86 (2010) 339–344

Adequate precautions to reduce an abnormally elevated maternaltemperature and to treat suspected infection are required to reducethe vulnerability of both the preterm and term brain from hypoxicinjury. On a practical level this has been made more difficult by therelatively common increase in body temperature associated withepidural analgesia. A temperature of 37.5 °C is commonplace in suchlabours and rather than promotewidespread use of antibiotics (whichis not recommended), a pyrexia needs to approach 38 °C beforetreatment is started unless other signs of sepsis exist. Thus para-cetamol is commonly used to keep temperatures down and this canthen mask what might otherwise have become a ‘clinical’ pyrexia.

5.2.2. Fetal heart rate monitoringMonitoring the fetal heart rate during labour is well established

but how best to do it remains the subject of research. When inter-mittent auscultation is used for low risk pregnancies twice as manybabies will require admission to neonatal intensive care with en-cephalopathy than if electronic monitoring is used [42] and yet thisis the practice supported by the Cochrane review and the NICEintrapartum guidelines [43] as there is no difference in mortality or incerebral palsy rates while operative intervention rates are higher withelectronic monitoring. The evidence to date is therefore confusingand often contradictory.

There have been some studies that suggest electronic fetal mon-itoring correlates well with metabolic acidosis and neurologic injury[44,45]. Williams and Galerneau [45] found that the best intrapartumfetal heart rate parameter to predict the development of significantacidemia was minimal/absent variability for at least 1 h as a solitaryabnormal finding or in conjunction with late decelerations in theabsence of accelerations. Although the sensitivity of these electronicfetal monitoring abnormalities for the identification of intrapartumasphyxia was 93% the positive predictive value was only 3%–18%.Studies on monkey fetuses [46] have shown that during the courseof progressive hypoxia that led to death, fetal heart rate variabilitydecreased and then disappeared, and late decelerations preceded fetalacidemia. It is generally believed that decreased short-term variabilityis the single most reliable sign of fetal compromise and normal short-term variability is a good indicator of normoxia. Computerised ante-natal fetal heart rate monitors are now available that identify ab-normalities in the short term variability of the fetal heart rate pattern.Their use in the intrapartum setting remains unclear and clinical trialsare underway.

Other studies have not found electronic fetal monitoring to beclinically helpful in the identification of fetal acidosis or neurologicinjury [47–50]. It has been suggested that given the low incidence ofneonatal encephalopathy the predictive value of an abnormal fetalheart rate tracing is poor. Other critics of electronic fetal monitoringpoint out that although fetuses with metabolic acidosis have sig-nificant episodes of bradycardia, decreased variability, nonreactivityand late decelerations the predictive power for HIE is not high.

There is evidence that changes in the ST segment of the electro-cardiogram (ECG) are related to fetal acidemia and that detection ofthese changes in combination with the cardiotocograph (CTG) canreduce the incidence of intrapartum asphyxia and/or neonatal en-cephalopathy [51–53]. Despite this initial optimism there remainreservations about this method of intrapartum monitoring [54] and anumber of problems have been raised since it was introduced intoroutine clinical practice in some units [55–57]. Other intrapartummonitoring techniques which have explored over the years includepulse oximetry and near-infrared spectroscopy, but neither have beenshown to have a place in clinical care [58,59].

On a practical level, the secret of intrapartum fetal monitoringremains one of vigilance with the midwife being present with thewoman throughout her labour to support her whilst midwife plus/minus obstetrician maintain a critical assimilation of signs and symp-toms in conjunction with an analysis of the fetal heart rate. Expecting

the fetal heart rate alone to give an answer is not logical and can helpexplain why the most complex methods of monitoring sometimesfail, as it can distract from other signs.

5.2.3. More liberal use of caesarean sectionThis is a very contentious suggestion because of the risks to both

mother and baby. Infants delivered by caesarean section have in-creased rates of respiratory problems compared to normally deliveredbabies. In addition, the surgical risks to the mother, particularly withrepeat caesarean sections are not inconsequential. Certainly in thepreterm situation the available evidence suggests that there is noreduction in neonatal mortality or incidence of cerebral abnormalitiesin babies delivered by caesarean section [60,61]. In the term infant thebest evidence for improved perinatal outcome is the term breech trial[62,63]. Perinatal mortality, neonatal mortality, or serious neonatalmorbidity was significantly lower for the planned caesarean sectiongroup (1.6%) than for the planned vaginal birth group (5%) (RelativeRisk 0·33 (95% CI 0·19–0·56)) but this benefit was no longer ap-parent at the two year follow-up [64].

5.2.4. Staff training, intrapartum guidelines and clinical governanceThe extent to which intrapartum asphyxia can be prevented by

improved care is controversial, but several investigations and malprac-tice claims analyses suggest that a significant proportion is preventable[65–69]. It often appears that substandard care is attributable to systemfailures frequently caused by poor communication between providers.

A recent study [70] suggested that suboptimal intrapartum careoccurred in 40–50% of cases with metabolic acidosis at birth. Mostcases of suboptimal care consisted of injudicious use of oxytocin or afailure to act on an abnormal fetal heart rate trace. The authors notedthat in many cases there appeared to be a gap between knowledgeand clinical practice and that availability of clinical intrapartum guide-lines did not reduce the incidence of adverse outcome. This trendis also supported by various national surveys in the United Kingdomsuggesting that suboptimal care was present in almost 70% of intra-partum fetal deaths.

Intrapartum care protocols for oxytocin usage, operative vaginaldelivery, shoulder dystocia, the use of magnesium sulphate, andcombined certification of physicians and nurses of training in fetalmonitoring can reduce malpractice claims paid, decrease caesareansection rates, and result in fewer adverse outcomes [71]. These are allnow included in standards of safety within maternity units and areaudited for CNST compliance.

In the United Kingdom, the Royal College of Obstetricians andGynaecologists recommend that any obstetric service delivery orga-nisation has a robust and transparent clinical governance frameworkwhich is applicable to each birth setting. Within this setting effectivemultidisciplinary care and good communication should be key factorsfor good practice. Furthermore, adequate staffing levels of all clinicaland support staff is required and this must be reviewed and auditedannually. Clinical leadership in promoting good practice and coordi-nating care is vital. Protocols based on clinical, organisational andsystem needs should also be available.

Professional staff should have the opportunity and support forcontinuing professional development, including agreed mandatorymultidisciplinary education and training sessions. Ongoing audit ofrelevant outcomes, with attention to any changes or trends is alsomandatory. These administrative factors are as important as clinicalinterventions in reducing the risk of hypoxic ischemic injury in thepreterm or term infant.

6. Conclusions

The initiation and evolution of brain injury in the term infant aftera hypoxic/ischemic insult is a very complex process with excitotoxi-city, oxidative stress and inflammation playing different roles within

343S. Kumar, S. Paterson-Brown / Early Human Development 86 (2010) 339–344

an environment of cerebral energy failure. Although there is now aneffective treatment in the form of neonatal brain cooling [72], manyaffected infants do not receive this intervention in time and for thosethat do receive it the response may not always be satisfactory. It isunlikely that, given the heterogeneity of risk factors, neonatal en-cephalopathy or hypoxic ischemic encephalopathy will be eradicated.The onus on obstetricians is therefore to minimise its occurrence withboth effective antenatal and preventative intrapartum strategieswhile refinements in neonatal treatment may optimise outcomes.Long-term follow-up of these infants is important to identify moresubtle disabilities such as learning difficulties or perceptual-motorproblems later in their life.

References

[1] de Haan M, Wyatt JS, Roth S, Vargha-Khadem F, Gadian D, Mishkin M. Brain andcognitive–behavioural development after asphyxia at term birth. Dev Sci 2006;9(4):350–8.

[2] Nelson KB, Leviton A. How much of neonatal encephalopathy is due to birthasphyxia? Am J Dis Child 1991;145(11):1325–31.

[3] Sarnat HB, Sarnat MS. Neonatal encephalopathy following fetal distress. A clinicaland electroencephalographic study. Arch Neurol 1976;33(10):696–705.

[4] Hankins GD, Speer M. Defining the pathogenesis and pathophysiology of neonatalencephalopathy and cerebral palsy. Obstet Gynecol 2003;102(3):628–36.

[5] Blair E, Stanley FJ. Intrapartum asphyxia: a rare cause of cerebral palsy. J Pediatr1988;112(4):515–9.

[6] Hagberg B, Hagberg G, Beckung E, Uvebrant P. Changing panorama of cerebralpalsy in Sweden. VIII. Prevalence and origin in the birth year period 1991–94. ActaPaediatr 2001;90(3):271–7.

[7] Thorngren-Jerneck K, Herbst A. Perinatal factors associated with cerebral palsy inchildren born in Sweden. Obstet Gynecol 2006;108(6):1499–505.

[8] Clark SL, Hankins GD. Temporal and demographic trends in cerebral palsy-fact andfiction. Am J Obstet Gynecol 2003;188(3):628–33.

[9] MacLennan A. A template for defining a causal relation between acute intrapartumevents and cerebral palsy: international consensus statement. Bmj 1999;319(7216):1054–9.

[10] ACOG committee opinion. Use and abuse of the Apgar score. Number 174–July 1996(replaces No. 49, November 1986). Committee on Obstetric Practice and AmericanAcademy of Pediatrics: Committee on Fetus and Newborn. American College ofObstetricians and Gynecologists. Int J Gynaecol Obstet 1996;54(3):303–5.

[11] American College of Obstetricians and Gynecologists (ACOG) and the AmericanAcademy of Pediatrics (AAP). Neonatal encephalopathy and cerebral palsy. Definingthe pathogenesis and pathophysiology. Am Coll Obstetricians Gynaecologists 2003.

[12] Graham EM, Ruis KA, Hartman AL, Northington FJ, Fox HE. A systematic review ofthe role of intrapartum hypoxia–ischemia in the causation of neonatalencephalopathy. Am J Obstet Gynecol 2008;199(6):587–95.

[13] Riley RJ, Johnson JW. Collecting and analyzing cord blood gases. Clin Obstet Gynecol1993;36(1):13–23.

[14] Goldaber KG, Gilstrap 3rd LC, Leveno KJ, Dax JS, McIntire DD. Pathologic fetalacidemia. Obstet Gynecol 1991;78(6):1103–7.

[15] Low JA, Lindsay BG, Derrick EJ. Threshold of metabolic acidosis associated withnewborn complications. Am J Obstet Gynecol 1997;177(6):1391–4.

[16] Sehdev HM, Stamilio DM, Macones GA, Graham E, Morgan MA. Predictive factorsfor neonatal morbidity in neonates with an umbilical arterial cord pH less than7.00. Am J Obstet Gynecol 1997;177(5):1030–4.

[17] Goodwin TM, Belai I, Hernandez P, Durand M, Paul RH. Asphyxial complications inthe term newborn with severe umbilical acidemia. Am J Obstet Gynecol 1992;167(6):1506–12.

[18] Jensen FE. Developmental factors regulating susceptibility to perinatal brain injuryand seizures. Curr Opin Pediatr 2006;18(6):628–33.

[19] Leviton A, Paneth N. White matter damage in preterm newborns—an epidemi-ologic perspective. Early Hum Dev 1990;24(1):1–22.

[20] Johnston MV, Trescher WH, Ishida A, Nakajima W. Neurobiology of hypoxic–ischemic injury in the developing brain. Pediatr Res 2001;49(6):735–41.

[21] Johnston MV, Hoon Jr AH. Possible mechanisms in infants for selective basalganglia damage from asphyxia, kernicterus, or mitochondrial encephalopathies. JChild Neurol 2000;15(9):588–91.

[22] Cowan F, Rutherford M, Groenendaal F, Eken P, Mercuri E, Bydder GM, et al. Originand timing of brain lesions in term infants with neonatal encephalopathy. Lancet2003;361(9359):736–42.

[23] Cowan F. Outcome after intrapartum asphyxia in term infants. Semin Neonatol2000;5(2):127–40.

[24] Maneru C, Serra-Grabulosa JM, Junque C, Salgado-Pineda P, Bargallo N, Olondo M,et al. Residual hippocampal atrophy in asphyxiated term neonates. J Neuroima-ging 2003;13(1):68–74.

[25] Okereafor A, Allsop J, Counsell SJ, Fitzpatrick J, Azzopardi D, Rutherford MA, et al.Patterns of brain injury in neonates exposed to perinatal sentinel events. Pediatrics2008;121(5):906–14.

[26] Rennie JM, Hagmann CF, Robertson NJ. Outcome after intrapartum hypoxicischaemia at term. Semin Fetal Neonatal Med 2007;12(5):398–407.

[27] Nelson KB, Lynch JK. Stroke in newborn infants. Lancet Neurol 2004;3(3):150–8.

[28] Wu YW, March WM, Croen LA, Grether JK, Escobar GJ, Newman TB. Perinatalstroke in children with motor impairment: a population-based study. Pediatrics2004;114(3):612–9.

[29] Mercuri E, Cowan F. Cerebral infarction in the newborn infant: review of theliterature and personal experience. Eur J Paediatr Neurol 1999;3(6):255–63.

[30] Gunther G, Junker R, Strater R, Schobess R, Kurnik K, Heller C, et al. Symptomaticischemic stroke in full-term neonates: role of acquired and genetic prothromboticrisk factors. Stroke 2000;31(10):2437–41.

[31] Lee J, Croen LA, Backstrand KH, Yoshida CK, Henning LH, Lindan C, et al. Maternaland infant characteristics associated with perinatal arterial stroke in the infant.Jama 2005;293(6):723–9.

[32] Himmelmann K, Hagberg G, Wiklund LM, Eek MN, Uvebrant P. Dyskinetic cerebralpalsy: a population-based study of children born between 1991 and 1998. DevMed Child Neurol 2007;49(4):246–51.

[33] Badawi N, Kurinczuk JJ, Keogh JM, Alessandri LM, O'Sullivan F, Burton PR, et al.Antepartum risk factors for newborn encephalopathy: the Western Australiancase–control study. Bmj 1998;317(7172):1549–53.

[34] Badawi N, Kurinczuk JJ, Keogh JM, Alessandri LM, O'Sullivan F, Burton PR, et al.Intrapartum risk factors for newborn encephalopathy: the Western Australiancase–control study. Bmj 1998;317(7172):1554–8.

[35] Sheldon RA, Sedik C, Ferriero DM. Strain-related brain injury in neonatal micesubjected to hypoxia–ischemia. Brain Res 1998;810(1–2):114–22.

[36] Yonekura I, Kawahara N, Nakatomi H, Furuya K, Kirino T. A model of globalcerebral ischemia in C57 BL/6 mice. J Cereb Blood Flow Metab 2004;24(2):151–8.

[37] Nunez JL, McCarthy MM. Sex differences and hormonal effects in a model ofpreterm infant brain injury. Ann N Y Acad Sci 2003;1008:281–4.

[38] National Institute for Clinical Excellence. Antenatal care: routine care for thehealthy pregnant woman CG62. NICE Press; 2008.

[39] Bricker L, Neilson JP, Dowswell T. Routine ultrasound in late pregnancy (after 24weeks' gestation). Cochrane Database Syst Rev 2008;4:CD001451.

[40] Dammann O, Leviton A. Role of the fetus in perinatal infection and neonatal braindamage. Curr Opin Pediatr 2000;12(2):99–104.

[41] Wu YW, Colford Jr JM. Chorioamnionitis as a risk factor for cerebral palsy: a meta-analysis. Jama 2000;284(11):1417–24.

[42] Alfirevic Z, Devane D, Gyte GM. Continuous cardiotocography (CTG) as a form ofelectronic fetal monitoring (EFM) for fetal assessment during labour. CochraneDatabase Syst Rev 2006;3:CD006066.

[43] National Institute for Clinical Excellence. Intrapartum care clinical guielines, 055.NICE Press; 2008.

[44] Low JA, Victory R, Derrick EJ. Predictive value of electronic fetal monitoring forintrapartumfetal asphyxiawithmetabolic acidosis.ObstetGynecol1999;93(2):285–91.

[45] Williams KP, Galerneau F. Intrapartum fetal heart rate patterns in the prediction ofneonatal acidemia. Am J Obstet Gynecol 2003;188(3):820–3.

[46] Murata Y, Martin Jr CB, Ikenoue T, Hashimoto T, Taira S, Sagawa T, et al. Fetal heart rateaccelerations and late decelerations during the course of intrauterine deathin chronically catheterized rhesusmonkeys. Am JObstetGynecol 1982;144(2):218–23.

[47] Williams KP, Galerneau F. Comparison of intrapartum fetal heart rate tracings inpatients with neonatal seizures vs. no seizures: what are the differences? J PerinatMed 2004;32(5):422–5.

[48] Samueloff A, Langer O, Berkus M, Field N, Xenakis E, Ridgway L. Is fetal heart ratevariability a good predictor of fetal outcome? Acta Obstet Gynecol Scand 1994;73(1):39–44.

[49] Ayres-de-Campos D, Costa-Santos C, Bernardes J. Prediction of neonatal state bycomputer analysis of fetal heart rate tracings: the antepartum arm of the SisPortomulticentre validation study. Eur J Obstet Gynecol Reprod Biol 2005;118(1):52–60.

[50] Spencer JA, Badawi N, Burton P, Keogh J, Pemberton P, Stanley F. The intrapartumCTG prior to neonatal encephalopathy at term: a case–control study. Br J ObstetGynaecol 1997;104(1):25–8.

[51] Amer-Wahlin I, Hellsten C, Noren H, Hagberg H, Herbst A, Kjellmer I, et al.Cardiotocography only versus cardiotocography plus ST analysis of fetal electrocar-diogram for intrapartum fetal monitoring: a Swedish randomised controlled trial.Lancet 2001;358(9281):534–8.

[52] Ojala K, Vaarasmaki M, Makikallio K, Valkama M, Tekay A. A comparison ofintrapartum automated fetal electrocardiography and conventional cardiotoco-graphy—a randomised controlled study. Bjog 2006;113(4):419–23.

[53] Westgate J, Harris M, Curnow JS, Greene KR. Plymouth randomized trial ofcardiotocogram only versus ST waveform plus cardiotocogram for intrapartummonitoring in 2400 cases. Am J Obstet Gynecol 1993;169(5):1151–60.

[54] Neilson JP. Fetal electrocardiogram (ECG) for fetal monitoring during labour.Cochrane Database Syst Rev 2006;3:CD000116.

[55] Melin M, Bonnevier A, Cardell M, Hogan L, Herbst A. Changes in the ST-intervalsegment of the fetal electrocardiogram in relation to acid-base status at birth. Bjog2008;115(13):1669–75.

[56] Westerhuis ME, Kwee A, van Ginkel AA, Drogtrop AP, Gyselaers WJ, Visser GH.Limitations of ST analysis in clinical practice: three cases of intrapartummetabolicacidosis. Bjog 2007;114(10):1194–201.

[57] Doria V, Papageorghiou AT, Gustafsson A, Ugwumadu A, Farrer K, Arulkumaran S.Review of the first 1502 cases of ECG-ST waveform analysis during labour in ateaching hospital. Bjog 2007;114(10):1202–7.

[58] East CE, Chan FY, Colditz PB, Begg LM. Fetal pulse oximetry for fetal assessment inlabour. Cochrane Database Syst Rev 2007;2:CD004075.

[59] Mozurkewich E, Wolf FM. Near-infrared spectroscopy for fetal assessment duringlabour. Cochrane Database Syst Rev 2000;3:CD002254.

[60] Hartling L, Chari R, FriesenC,VandermeerB, Lacaze-Masmonteil T. A systematic reviewof intentional delivery in women with preterm prelabor rupture of membranes.J Matern Fetal Neonatal Med 2006;19(3):177–87.

344 S. Kumar, S. Paterson-Brown / Early Human Development 86 (2010) 339–344

[61] Arpino C, Brescianini S, Ticconi C, Di Paolo A, D'Argenzio L, Piccione E, et al. Doescesarean section prevent mortality and cerebral ultrasound abnormalities inpreterm newborns? J Matern Fetal Neonatal Med 2007;20(2):151–9.

[62] Su M, Hannah WJ, Willan A, Ross S, Hannah ME. Planned caesarean sectiondecreases the risk of adverse perinatal outcome due to both labour and deliverycomplications in the Term Breech Trial. Bjog 2004;111(10):1065–74.

[63] Hannah ME, Hannah WJ, Hewson SA, Hodnett ED, Saigal S, Willan AR. Plannedcaesarean section versus planned vaginal birth for breech presentation at term: arandomisedmulticentre trial. TermBreech Trial Collaborative Group. Lancet 2000;356(9239):1375–83.

[64] WhyteH, HannahME, Saigal S, HannahWJ, Hewson S, Amankwah K, et al. Outcomesof children at 2 years after planned cesarean birth versus planned vaginal birth forbreech presentation at term: the International Randomized Term Breech Trial. Am JObstet Gynecol 2004;191(3):864–71.

[65] Draper ES, Kurinczuk JJ, Lamming CR, Clarke M, James D, Field D. A confidentialenquiry into cases of neonatal encephalopathy. Arch Dis Child Fetal Neonatal Ed2002;87(3):F176–80.

[66] Holt J, Fagerli I, Holdo B, Vold IN. Audit of neonatal deaths of nonmalformed infantsof 34 or more weeks' gestation: unavoidable catastrophic events or suboptimalcare? Acta Obstet Gynecol Scand 2002;81(10):899–904.

[67] Becher JC, Stenson BJ, Lyon AJ. Is intrapartum asphyxia preventable? Bjog 2007;114(11):1442–4.

[68] Berglund S, Grunewald C, Pettersson H, Cnattingius S. Severe asphyxia due todelivery-related malpractice in Sweden 1990–2005. Bjog 2008;115(3):316–23.

[69] Jonsson M, Norden SL, Hanson U. Analysis of malpractice claims with a focus onoxytocin use in labour. Acta Obstet Gynecol Scand 2007;86(3):315–9.

[70] Jonsson M, Norden-Lindeberg S, Ostlund I, Hanson U. Metabolic acidosis at birthand suboptimal care—illustration of the gap between knowledge and clinicalpractice. Bjog 2009;116(11):1453–60.

[71] Clark SL, Belfort MA, Byrum SL, Meyers JA, Perlin JB. Improved outcomes, fewercesarean deliveries, and reduced litigation: results of a new paradigm in patientsafety. Am J Obstet Gynecol 2008;199(2):105 e1–7.

[72] Azzopardi DV, StrohmB, Edwards AD, Dyet L, Halliday HL, Juszczak E, et al. Moderatehypothermia to treat perinatal asphyxial encephalopathy. N Engl J Med 2009;361(14):1349–58.