Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                

Current Concepts in Intrauterine Growth Restriction

2004, Journal of Intensive Care Medicine

Current Concepts in Intrauterine Growth Restriction Dara Brodsky, MD Helen Christou, MD Regulation of fetal growth is multifactorial and complex. Diverse factors, including intrinsic fetal conditions as well as maternal and environmental factors, can lead to intrauterine growth restriction (IUGR). The interaction of these factors governs the partitioning of nutrients and rate of fetal cellular proliferation and maturation. Although IUGR is probably a physiologic adaptive response to various stimuli, it is associated with distinct short- and long-term morbidities. Immediate morbidities include those associated with prematurity and inadequate nutrient reserve, while childhood morbidities relate to impaired maturation and disrupted organ development. Potential long-term effects of IUGR are debated and explained by the fetal programming hypothesis. In formulating a comprehensive approach to the management and follow-up of the growth-restricted fetus and infant, physicians should take into consideration the etiology, timing, and severity of IUGR. In addition, they should be cognizant of the immediate perinatal response of the growth-restricted infant as well as the childhood and longterm associated morbidities. A multidisciplinary approach is imperative, including early recognition and obstetrical management of IUGR, assessment of the growth-restricted newborn in the delivery room, possible monitoring in the neonatal intensive care unit, and appropriate pediatric follow-up. Future research is necessary to establish effective preventive, diagnostic, and therapeutic strategies for IUGR, perhaps affecting the health of future generations. Key words: intrauterine growth restriction; small for gestational age Definition and Classification Intrauterine growth restriction (IUGR) describes a decrease in fetal growth rate that prevents an infant From Children’s Hospital, Harvard Medical School, Department of Newborn Medicine, Boston, Massachusetts. Received Dec 15, 2003. Received revised Mar 8, 2004. Accepted for publication Mar 10, 2004. Address correspondence to Dara Brodsky, MD, Children’s Hospital, Harvard Medical School, Department of Newborn Medicine, Enders 9, 300 Longwood Avenue, Boston, MA 02115, or e-mail: dara.brodsky@tch.harvard.edu. Brodsky D, Christou H. Current concepts in intrauterine growth restriction. J Intensive Care Med. 2004;19:000-000. DOI: 10.1177/0885066604269663 Copyright © 2004 Sage Publications from obtaining his or her complete growth potential. IUGR infants are small for gestational age (SGA) if their birth weight measures less than 3% to 10% using standard growth curves [1]. Less commonly, these infants may be characterized as appropriate for gestational age (AGA) if their growth restriction is mild. It is important to distinguish between infants who experienced in utero growth restriction from infants with normal in utero growth but constitutionally small (ie, no loss in percentiles throughout gestation) since IUGR fetuses undergo fetal adaptations to a pathologic condition, leading to distinct short- and long-term outcomes. Two main patterns of fetal growth restriction are observed (see Table 1). If fetal growth is impaired during the first or second trimester, the infant will have symmetric growth restriction. This proportional lack of growth is caused by reduced fetal cellular proliferation of all organs and occurs in approximately 20% to 30% of IUGR infants [2]. In contrast, asymmetric growth in which an infant has a smaller abdominal size compared to head size will occur if the decrease in growth velocity happens in the last trimester. This head-sparing phenomenon is the most common form of IUGR (~70%-80%) [2] and is attributed to the ability of the fetus to adapt, redistributing its cardiac output to the spleen, adrenal, coronary, and cerebral circulations. Although some overlap can occur, the timing of the growth delay is more important than the etiology in determining the pattern of growth restriction. Epidemiology and Etiology The incidence of IUGR is estimated to be approximately 5% to 7%. Some studies identify a greater percentage (up to 15% of pregnancies), but these reports define IUGR and SGA as equivalent. Despite advances in obstetric care, IUGR remains prevalent in developed countries. However, the causes of IUGR in these areas are different than in 1 Brodsky, Christou Table 1. Specific Distinctions Between Symmetric and Asymmetric Intrauterine Growth Restriction (IUGR) Symmetric IUGR Incidence Period of growth restriction Physical characteristics Pathophysiology Etiology Outcome Asymmetric IUGR 20%-30% Begins first or second trimester Small head and abdominal size Impaired cellular embryonic division Impaired cellular hyperplasia ± hypertrophy Decreased cell number ± size Mostly intrinsic: chromosomal abnormalities and congenital malformations Drugs Infection Early-onset severe preeclampsia Preeclampsia <30 wk superimposed with chronic hypertension Greater morbidity and mortality 70%-80% Begins third trimester Large head size relative to small abdomen Impaired cellular hypertrophy Decreased cell size Mostly extrinsic: placental and maternal vascular factors (eg, placental insufficiency) Lower morbidity and mortality This table outlines the differences between symmetric and asymmetric IUGR. Although most infants can be categorized into one of these groups, some infants may have features of both types of IUGR [2,8]. Table 2. Maternal, Placental, and Fetal Etiologies of Intrauterine Growth Restriction (IUGR) Maternal Vascular disorders (~25%-30%) Hypertension Diabetes mellitus Renal disease Collagen vascular disease Hypercoaguable states Thrombophilia Antiphosopholipid antibody syndrome Persistent hypoxia (eg, high altitude, pulmonary or cardiac disease, severe anemia) Undernutrition Toxins (eg, tobacco, alcohol, medications, and illicit drugs, irradiation) Uterine malformation or masses Placental Fetal Abnormal trophoblast invasion Genetic (~20%) Placental infarcts Chromosomal abnormalities Placenta previa Syndromes/congenital malformations Circumvallate placenta Multiple gestation (5%) Chorioangiomata Intrauterine infection Velamentous umbilical cord insertion Cytomegalovirus Umbilical-placental vascular anomalies Malaria Parvovirus Rubella Toxoplasmosis Herpes virus HIV Modified from Lee et al [19], Schwart [14], and Lin and Santolaya-Forgas [8]. Overall, IUGR encompasses an extremely heterogeneous group that can be categorized into maternal, placental, and fetal factors. There is a subgroup of infants with overlapping etiologies. In addition, there are also a significant number of infants with unexplained etiologies. Third World nations. In most Western societies, placental insufficiency is the major cause of IUGR, while inadequate maternal nutrition and malaria infections play a greater role in developing countries [3,4]. Overall, IUGR encompasses an extremely heterogeneous group. While a large number of etiologies are not identified, the known associations involve fetal, placental, and/or maternal factors (see Table 2). There is a strong link between IUGR, chromosomal abnormalities, and congenital malformations. Specifically, fetuses with chromosomal disorders such as trisomy 13, 18, and 21 often have impaired growth. Also, fetuses with other autosomal irregularities (eg, deletions, ring chromosomes) typically have inadequate growth [5]. Indeed, of 458 fetuses 2 17 to 39 weeks gestation with IUGR, 19% (89) had chromosomal abnormalities (most commonly trisomy 18) [6]. Among 13,000 infants born with major malformations (most commonly anencephaly), 22% experienced IUGR [7]. Less frequently, IUGR may be due to first or second trimester fetal infection, including cytomegalovirus, malaria, parvovirus, and rubella. The majority of fetal etiologies lead to early gestation symmetric IUGR. Chronic maternal vascular disease due to hypertension, diabetes mellitus, renal disease, or collagen vascular disease is the most common cause of IUGR [8] in developed countries. The most profound effect is observed if the hypertension is early onset, severe, or due to chronic hypertension with superimposed preeclampsia [5]. Hypercoaguable Journal of Intensive Care Medicine 18(X); 2004 Intrauterine Growth Restriction maternal conditions such as thrombophilia and antiphospholipid antibody syndrome also inhibit growth either by placental thrombosis formation or by secondary effects of maternal hypertension [9,10]. Persistent maternal hypoxia due to high altitude, severe pulmonary or cardiac disease, and/or severe chronic anemia limits oxygen delivery to the fetus and attenuates fetal growth. Periods of famine in the Netherlands, Germany, and the former Soviet Union have shown that severe maternal malnutrition can also impair fetal growth [11,12]. The severity of the available food supply [13] and the length of malnutrition [12] correlate with the degree of growth delay. While the fetus is affected by chronic severe maternal malnutrition, it seems to be fairly resistant to acute malnutrition, particularly if it occurs late in gestation [14]. Maternal toxins can contribute to the development of a growth-restricted fetus. Cigarette smoking reduces uterine blood flow, limiting fetal oxygenation and attenuating growth [15]. The quantity of cigarettes smoked per day correlates with the degree of IUGR [16-18]. Prolonged maternal alcohol ingestion and other drugs (eg, steroids, coumadin, hydantoin, cocaine, and heroin) are also implicated in the development of IUGR [19]. In addition to these factors, physical constraints such as large placental abnormalities, uterine masses, or multiple gestations can lead to growth-restricted fetuses. Pathophysiology These multifactorial causes of IUGR create 3 possible scenarios: (1) abnormal placental function, (2) inadequate maternal supply of oxygen and/or nutrients, and/or (3) decreased ability of the fetus to use the supply (Fig 1). The placenta plays an integral role in the first 2 categories. Abnormal development, inadequate perfusion, and dysfunction of the placental villi are often responsible for the development of IUGR, especially the earlyonset type. Indeed, placentas from mothers with preeclampsia are characterized by shallow cytotrophoblast (CTB) invasion of the uterus and abnormal CTB differentiation [20]. Inadequate CTB invasion will prevent effective destruction of distal spiral decidual arteries leading to inadequate maternal perfusion of placental villi, local placental hypoxia, and impaired fetal growth [21]. In addition, increased apoptosis of trophoblasts creates maldevelopment of placental villi, preventing maximal attainment of placental function [22]. Oxidative Journal of Intensive Care Medicine 18(X); 2004 stress, infarction, cytokine damage, and hypertension can further inhibit optimal function of placental villi [22-24]. The response of the placental villi to damage from ischemia and/or hypoxia may involve erratic angiogenesis, limiting the possibility of complete placental recovery [22]. Recently, some specific molecular pathways have been associated with the development of IUGR. Since insulin and its associated insulin-like growth factor (IGF)-I and –II are the primary anabolic hormones necessary for fetal growth, it is postulated that they may play a critical role in the development of IUGR [25,26]. Indeed, low levels of IGF-1 and elevated levels of IGF binding protein (BP)-1 have consistently been observed in growth-restricted infants [27-29]. The involvement of IGF is directly supported by a report of a patient with a homozygous partial deletion of the IGF-I gene who had extreme prenatal growth restriction that continued postnatally [30]. In addition, IGF-1 receptor mutations have been identified in 2 children with poor intrauterine and postnatal growth [31]. Animal studies also support the role of the IGF signaling pathway in the development of IUGR. Transgenic fetal mice overexpressing IGFBP-1 demonstrate impaired fetal growth [32]. In addition, newborn mice homozygous null for the IGF-1, IGF-2, and IGF-1 receptor genes demonstrate growth deficiency with birth weights 45% to 60% of normal size [25,33]. Since transgenic mice with excess maternal decidual IGFBP-1 have abnormal trophoblast invasion and differentiation, the IGF-1 pathway may also regulate placental development [32]. Moreover, placental-specific IGF-2 knockout mice develop IUGR, emphasizing the important role of the placenta in fetal growth restriction. In addition to fetal growth and placental development, it has been suggested that this complex IGF hormonal system also regulates maternal metabolism [34-37]. Another potential signaling pathway involved in placental causes of IUGR is the glial cell missing–1 (GCM1) gene, which is necessary for trophoblast morphogenesis and differentiation [38,39]. Targeted disruption of this transcription factor in trophoblasts leads to inadequate trophoblast differentiation and similar placental histology as that found in early-onset IUGR fetuses [39,40]. Perhaps the gene itself or signaling pathways leading to GCM1 expression are altered, producing maldevelopment of the placenta villi. Accumulating evidence from animal and human studies supports that leptin may also participate in the regulation of fetal growth and development [41]. Alterations in the expression or activity of placental transporters, particularly those that transfer 3 Brodsky, Christou Fig 1. Pathophysiology of intrauterine growth restriction (IUGR). The potential factors leading to IUGR influence the maternal supply to the fetus, the ability of the fetus to use the maternal supply, and/or the placental function. If IUGR develops, there are numerous fetal adaptations that may incur perinatal, childhood, and adult effects. RDS = respiratory distress syndrome; NEC = necrotizing enterocolitis; IVH = intraventricular hemorrhage; SIDS = sudden infant death syndrome. amino acids, may also contribute to the development of IUGR [42]. Animal models have shown that pregnant rats with protein malnutrition had growth-restricted fetuses associated with a decrease in the Na+-dependent system A amino acid transporter [43]. It is possible that the placenta senses its own nutrient supply, alters its transport abilities accordingly, and regulates fetal growth; these mechanisms have not yet been well defined. 4 Prenatal Management of Growth-Restricted Fetus It is crucial that the obstetrician recognizes and accurately diagnoses a fetus with IUGR. Currently, the recommended method is by measuring anthropometric parameters, which include fetal abdominal circumference, head circumference, biparietal Journal of Intensive Care Medicine 18(X); 2004 Intrauterine Growth Restriction diameter, and length. These results are converted to fetal weight estimates using standard formulas and compared with population-based fetal growth curves at specific gestational ages (GA) [5]. Precise initial dating by an early ultrasound will ensure that the accurate GA is used [44]. Sequential assessments are necessary to determine if there is a decrease in the fetal growth rate. Once the diagnosis of IUGR is established, the obstetrician must attempt to determine the specific cause of the growth delay. An abnormal fetal survey should prompt further investigations, including fetal karyotyping, specialized genetic tests, and possible consultation with a geneticist. If a viral infection is suspected, maternal serum studies are required. Since fetal growth restriction may be the first evidence of preelampsia, maternal blood pressures should be monitored closely. The pregnant woman should be evaluated for vascular and hypercoaguable diseases as well as hypoxemic conditions. An assessment for malnutrition and medications or toxins, including tobacco, may be helpful. After delivery, examination of the placenta by a pathologist may disclose placental vascular abnormalities. Despite numerous approaches to managing IUGR, there are no effective therapies to improve the growth pattern of a fetus [45]. Modalities tested include maternal nutritional supplementation, plasma volume expansion, and maternal medications such as low-dose aspirin [5,46,47]. Although maternal oxygen supplementation has been shown to increase fetal oxygen tension [48] and transiently improve fetal well-being [49,50], long-term benefits have not been demonstrated [51]. Some reports suggest that any short-term benefit of improved fetal oxygenation may actually worsen the longterm outcome by prolonging the time a fetus spends in an inadequate intrauterine environment [52]. Although antihypertensive medications can be advantageous in the treatment of preeclampsia, they do not prevent or reverse fetal growth restriction [8]. Finally, while bed rest is often encouraged, there is no evidence to support that this approach has any effect on fetal growth [53]. Since there are no effective therapies to reverse fetal growth restriction, prenatal management is aimed primarily at determining the ideal timing and mode of delivery. This assessment must be individualized, depending on several variables: GA of the fetus, maternal health, severity of the IUGR, and fetal well-being. Perhaps optimizing the delivery time and removing the fetus from a suboptimal environment can prevent the risk of hypoxia and significant morbidities. Journal of Intensive Care Medicine 18(X); 2004 The GA of the fetus is a critical component of the decision-making process. If a growth-restricted fetus is near term with either poor growth or associated severe maternal preeclampsia, delivery is recommended. Similarly, an IUGR fetus between 34 and 37 weeks gestation should be delivered if there are similar concerns in the setting of mature fetal lung indices. The management of premature infants <34 weeks gestation is more challenging. Indeed, arbitrary delivery of a growth-restricted premature fetus at any GA is not warranted because fetal mortality is lower than neonatal mortality prior to 31 weeks gestation [54]. Since antenatal steroids decrease neonatal pulmonary and central nervous system morbidity in premature infants, these are still recommended for IUGR premature fetuses, although specific effects on fetuses with growth delay are conflicting [55,56]. In the group of IUGR fetuses with unclear indications for immediate delivery, the obstetrician must rely on other assessment tools to monitor fetal well-being and determine the safest time for delivery. While some physicians advocate biweekly nonstress tests, randomized controlled studies have not shown that outcomes are improved with this monitoring [57,58]. Similarly, trials assessing efficacy of using biophysical profiles as a modality to determine the optimal delivery time have not shown an association with better outcomes [47,54,57]. Since studies demonstrate that oligohydramnios dramatically increases perinatal mortality in the growth-restricted fetus, this is often used as an independent indicator for delivery [59]. Umbilical Doppler flow measurements are the most valuable current technique to distinguish the sick IUGR fetus from the well IUGR fetus [60] and, in contrast to other modalities, improve perinatal outcomes [61-64]. Indeed, using this approach, fetuses with the worst placental outcome [65,66] and the most detrimental perinatal outcome [67-69] can be identified. During this technique, the impedance to blood flow in fetal arteries and veins by Doppler velocimetry is measured; absent or reversed end-diastolic flow in the umbilical artery indicates an inadequate fetal status [70,71], while normal umbilical Doppler flow is rarely associated with significant morbidity [72]. If associated abnormalities are observed in the ductus venosus and umbilical veins, the IUGR fetus is even more compromised with a high chance of imminent death [73]. This corresponds with the timing of fetal circulatory changes during a compromised state since venous alterations typically occur after arterial changes [74]. The assessment of fetal acid-base balance by umbilical blood sampling has been shown 5 Brodsky, Christou to further identify growth-restricted fetuses with hypoxia and acidosis [75]. Some new modalities are currently being investigated to improve the assessment of a compromised fetus. One method records Doppler fetal cerebral circulation and calculates the cerebro-placental ratio of blood flow [76]. While this ratio has been shown to be impaired in IUGR fetuses, a correlation between perinatal complications and neurodevelopmental outcome has not been deciphered [77,78]. Qualitative determination of general movements is also proposed as a potential tool to assess brain function [79-81]. Indeed, preliminary findings have found that fidgety movements are predictive of neurological insults [82]. Finally, Doppler imaging of the proximal uterine arteries to assess maternal vascular function has been helpful in predicting the severity of IUGR in mothers with preeclampsia [83]. Once the timing of delivery has been established, the obstetrician is now responsible for determining the mode of delivery. A trial of labor is often considered in selected women with growth-restricted fetuses if antepartum fetal testing by umbilical artery Doppler velocimetry is normal [84]. Despite this preselection of milder IUGR fetuses, a cesarean delivery may still be required since all growthrestricted fetuses have a greater rate of intolerance to labor, evident by abnormal fetal heart rate patterns [85]. Management and Outcomes During the Neonatal Period Numerous studies have shown that growthrestricted fetuses have a greater risk of fetal mortality, correlating directly with the severity of IUGR [5,54,86-90]. Indeed, studies have found that almost half of stillborn fetuses without malformations had growth restriction [89]. This increased mortality risk extends beyond the fetal period; growth-restricted full-term infants weighing 1500 to 2500 g (<10%) have a 5 to 30 times higher mortality risk compared to term infants born >2500 g (>10%) [5]. It is well recognized that growth-restricted infants are at greater risk of morbidities [86,88,90,91]. Neonates with a greater severity and protracted period of IUGR are at even more risk of perinatal complications and poor prognoses. It is also important to acknowledge that known etiologies of IUGR will independently influence these morbidities. Specifically, infants with IUGR attributable to fetal factors (eg, in utero infection, chromosomal abnormalities) have independent adverse outcomes. 6 At delivery, the presence of a qualified neonatology team is an important initial step in managing IUGR infants. Infants may have perinatal depression with low APGAR scores requiring emergent resuscitation in the delivery room [5,92,93]. This may be associated with meconium aspiration and/or a severe metabolic acidosis. Because of decreased liver glycogen and fat stores, IUGR infants must be monitored for hypoglycemia and hypothermia [94]. Polycythemia attributed to a relatively hypoxic intrauterine environment may predispose the IUGR infant to greater postnatal hemolysis and lead to indirect hyperbilirubinemia. In addition, since this population of infants is at increased risk of intrauterine coagulative processes, thrombocytopenia can be observed [95]. The high incidence of premature delivery in IUGR infants leads to the characteristic postnatal risks of prematurity [90,96]. Numerous investigations have compared specific complications of prematurity among growth-restricted neonates and gestational age–matched AGA infants. For almost 3 decades, it was believed that IUGR infants had accelerated lung maturation with decreased rates of hyaline membrane disease (HMD) due to prolonged intrauterine stress inducing fetal corticosteroid production. However, recent studies suggest that HMD occurs at equal rates in growth-restricted and AGA premature infants [97-99] and perhaps may actually be more common in IUGR infants [44,90]. Most trials have demonstrated an increased risk of chronic lung disease in IUGR infants [97,98,100]. Perhaps the restricted development of the fetus enhances the lung’s susceptibility to postnatal injury. Similar to HMD, the association of necrotizing enterocolitis (NEC) and poor intrauterine growth remains controversial. It has generally been believed that the decreased vascular supply to the gastrointestinal tract in utero predisposes the infant to develop NEC, and studies have demonstrated this increased risk [44,90,99,101,102]. However, other reports have shown no increased risk of NEC in IUGR infants [103]. Some reports demonstrate that prenatal findings of abnormal flow in the fetal aorta as well as absent or reversed end-diastolic umbilical arterial flow predisposes the growthrestricted fetus to develop NEC [104,105]. Further studies are needed to elucidate the correlation of NEC and IUGR. In addition to HMD and NEC, the association of intraventricular hemorrhage (IVH) in growthrestricted and AGA premature infants is still debated. Some studies have demonstrated a larger risk of IVH [90], while others did not observe any associaJournal of Intensive Care Medicine 18(X); 2004 Intrauterine Growth Restriction tion [99,101]. Another report found that IVH was actually decreased in IUGR infants of 28 weeks gestational age compared to AGA neonates, suggesting a potential protective effect of growth restriction [44]. The heterogeneity of IUGR infants may contribute to the discrepancies in potential associations with HMD, NEC, and IVH. Although there is a low number of published investigations focused on hypothyroxinemia and IUGR, all have been consistent; growth-restricted infants are at greater risk for early hypothyroxinemia [106-109]. Metabolic reasons for this association have not yet been identified. A higher infection rate has also been consistently observed in IUGR premature infants compared with matched AGA infants [99,110]. An increased association between persistent pulmonary hypertension of the newborn (PPHN) and asymmetric growth restriction has also been reported [111]. Our anecdotal experience suggests that premature IUGR infants are a subset that is more likely to develop PPHN. Finally, the kidneys appear to be extremely vulnerable to IUGR and are often small in proportion to body weight [112,113]. Besides the deficit in nephrons, there is an impairment of renal function associated with IUGR [114,115]. These renal effects may explain the epidemiological studies connecting the development of adult hypertension with IUGR [116-118]. Management and Outcomes During Childhood The effects of IUGR persist beyond the neonatal period and may have profound influences during childhood. The majority of IUGR infants demonstrate an increased postnatal growth velocity with catch-up growth by 2 to 3 years of age [19,119,120]. However, since IUGR infants have low nutritional stores and often have feeding difficulties, a subgroup (~10%) remains vulnerable to continued growth problems [19,121]. Longitudinal studies support a greater incidence of short stature in IUGR children born prematurely compared with full-term IUGR children [120,122]. If a child with a history of IUGR is older than 3 years and has persistent growth delay, he or she should still be evaluated by a pediatric endocrinologist for other potential causes [19]. In addition to growth hormone deficiency, one should assess for abnormalities of IGF-1, IGFBP-3, fasting insulin, glucose, and lipid levels [19]. Some studies demonstrate normalization of height after prolonged growth hormone treatment despite normal initial hormonal levels [123-131]. Journal of Intensive Care Medicine 18(X); 2004 The literature is inconsistent about the influence of IUGR on long-term neurological outcome, ranging from minimal decreases in IQ [132,133] or no effects [134-138] to cerebral palsy [139-141]. Not surprisingly, the worst outcomes are observed in those with severe IUGR [134], early-onset IGUR [142,143], premature birth [144], and prenatal evidence of impaired umbilical arterial flow [145,146]. The presence of microcephaly at birth with poor catch-up of head growth is one of the most significant negative determinants of neurodevelopmental outcome [81,147]. The best long-term neurological prognosis is encountered when the fetus tolerates labor, the growth restriction is mild, the delivery date is carefully determined, immediate neonatal care is available, and there is appropriate prenatal and postnatal head growth [2,5,147]. Most reports suggest that severe IUGR incurs subtle behavioral and learning disabilities during childhood [77,135,144,148-151]. Perhaps the disparity of findings related to neurological outcome can be explained by the heterogeneity of the populations examined including differences in etiology, definition of IUGR, age the children are examined, perinatal and neonatal complications, and distinct sociodemographic and postnatal environmental factors [35]. Other childhood effects from IUGR have been identified. There is a slight increased risk of sudden infant death syndrome reported in growth-restricted infants [152,153], attributed possibly to failure of breathing or arousal from sleep [154,155]. Fetal growth restriction also impairs airway function during childhood, evident by lower forced expiratory volumes in 5- to 11-year-old children with a history of IUGR [156]. The onset duration and severity of IUGR may influence these childhood effects. Long-term Outcomes It is well accepted that an individual’s risk of developing adult disease is determined by the interactions between one’s genetic makeup and postnatal environmental factors. Epidemiological studies since the early 1980s suggest that suboptimal growth during the fetal period also predisposes to specific diseases later in life including adultonset hypertension (HTN), type 2 diabetes, and coronary artery disease (CAD) [157]. This epidemiological association forms the basis of the fetal programming hypothesis, which proposes that these 3 disorders “originate in developmental plasticity, in response to undernutrition during fetal life and infancy” [158]. 7 Brodsky, Christou Although the association of IUGR and HTN in adulthood is still disputed [159], accumulating evidence from animal and human studies supports that suboptimal in utero nutrient supply leads to metabolic and hormonal adaptations as well as altered organ structure, which may contribute to HTN, insulin resistance, and CAD. The so-called “thrifty phenotype” describes the fetal response to inadequate nutrient supply and consists of decreased muscle mass, insulin resistance, decreased capillary network, and an increased stress response. In the setting of this phenotype, it has been proposed that subsequent abundance of nutritional supplements may predispose an individual to the development of the metabolic syndrome (syndrome X), which consists of centripetal fat accumulation, insulin resistance, glucose intolerance, hypertriglyceridemia, low levels of high-density lipids, and HTN. Since this syndrome is complex and multifactorial, the fetal programming hypothesis can be viewed as the result of the interaction between the genetic makeup of the individual and nutritional, metabolic, and hormonal cues during fetal and neonatal development that ultimately determine the risk of adult morbidities. Several studies have addressed the possible association of IUGR with HTN in childhood and adulthood [157,160]. Although the majority of these studies support an inverse relationship between birth weight and arterial blood pressure, a published meta-analysis suggests that as the statistical size of the study increases, the magnitude of this association decreases [159]. In evaluating these studies, several methodological considerations should be taken into account, including the source of the birth weight (birth records or memory), the number of blood pressure measurements, the age at which blood pressure is reported, and whether there is adjustment for current weight and other confounding variables. Animal and human studies have provided some insight into the possible biological basis for the association of IUGR and HTN. Because IUGR infants have a significant reduction in nephron number [114,161,162], this may predispose them to develop HTN later in life [116-118,163]. Furthermore, since the etiology of HTN is multifactorial and largely unknown, it is possible that IUGR may be a modifying factor in individuals who have a genetic predisposition to HTN. Clearly, further elucidation of the underlying molecular mechanisms is needed to accomplish early detection and management of at-risk infants. Many epidemiological studies support a strong association between IUGR and the subsequent 8 development of type 2 diabetes and the metabolic syndrome [164,165]. Animal data show that this association can be explained partly by permanent changes in the expression of hormone receptors, signaling molecules, and regulatory enzymes in liver, pancreatic β cells, muscle, and adipose tissue in response to in utero undernutrition [165-167]. These studies have also questioned whether rapid catch-up growth following early growth retardation may add to the risk of developing insulin resistance [165]. The combination of low birth weight and accelerated childhood growth is associated with exaggeration of adiposity and insulin resistance, particularly for those who were born with IUGR. The postnatal age of adiposity rebound is a predictor of both adult adiposity and insulin resistance. In one study, individuals with adiposity rebound before age 5 had an 8.6% incidence of diabetes compared to 2.1% in individuals whose adiposity rebound occurred after age 7 [168]. Recent studies in premature human neonates suggest that enhanced nutritional intake postnatally is associated with an increased incidence of insulin resistance at age 16 [169]. Finally, it has been proposed that IUGR is an added risk factor in individuals who have another predisposing factor for insulin resistance such as PPAR-γ polymorphisms [170]. The association between IUGR and CAD is predominantly indirect, mediated by the effect of low birth weight on the incidence of HTN, diabetes, and the metabolic syndrome. Studies of in utero anemia in sheep have provided evidence for a direct effect of intrauterine anemia (and the associated growth restriction) on coronary artery physiology. Altered coronary conductance and inappropriate cardiac responses to hypoxic stress have been observed in adult sheep exposed to in utero anemia [171,172]. The importance of these observations in humans needs to be clarified. In summary, IUGR has been associated with specific adult diseases. It appears that the nutritional management of the low-birth-weight infant probably plays a role in the risk of developing some long-term morbidities. Regular monitoring of weight, height, and body mass index during infancy, childhood, and adolescence should be implemented to attempt to avoid excessive weight gain and possibly HTN, type 2 diabetes, and CAD. Future Directions Innovative intrauterine interventions to prevent, detect, and treat growth restriction could reduce potential neonatal, childhood, and adult effects. Journal of Intensive Care Medicine 18(X); 2004 Intrauterine Growth Restriction Presently, there are 3 effective preventive strategies to optimize fetal growth: (1) discourage smoking during pregnancy, (2) encourage optimal maternal nutrition, and (3) prescribe antimalarial prophylaxis for primigravida in developing countries [45]. Future research necessitates the development of other approaches to prevent IUGR. Since the delay in fetal growth is often insidious, we need to develop new, accurate diagnostic tools to identify fetuses at the early stages of IUGR. Doppler ultrasounds may become more sensitive to subtle vascular changes. It is also possible that ultrasound imaging could measure fat concentrations during late gestation to decipher small, wellproportioned fetuses with appropriate growth velocities from IUGR fetuses [173]. Currently, research trials are investigating the ability to quantify placental transfer by administering a cocktail of stable isotopes (eg, glucose, specific amino acids) to pregnant mothers. Since these substrates are expected to cross the placenta at a standard rate, measurement of fetal concentrations by cordocentesis would correlate with placental function [35,174]. New fetal diagnostic techniques to assess fetal well-being are critical to guide obstetricians about the optimal timing and mode of delivery. Ideally, these approaches would correlate with short- and long-term outcomes. Future studies will need to identify effective therapeutic strategies that could reverse fetal growth restriction. Targeting the anabolic hormone IGF-1 may be one possible approach. Infusion of IGF-1 into fetal sheep increased growth of the heart, liver, kidneys, spleen, and pituitary gland [175]. Since this may not be feasible in humans, maternal administration of IGF-1 may improve fetal growth. Unfortunately, one study demonstrated that pregnant rats who received IGF-1 had accelerated growth of maternal tissues without any changes in fetal growth [37]. Further research focused on IGF1 as a therapy for IUGR is required. A second approach could focus on enhancing placental growth and/or function to potentially induce fetal growth. To accomplish these therapeutic goals, the distinct pathophysiology of IUGR must be elucidated using additional molecular techniques, animal models, epidemiological studies, and clinical investigations. These tools must unravel the intricate interactions between the health of the pregnant woman, the function of the placenta, and the growth of the fetus. This integrative approach requires tremendous consistency in the definition, classification, etiology, and severity of IUGR. It is hoped that these objectives will enhance our abiliJournal of Intensive Care Medicine 18(X); 2004 ty to establish effective preventive, diagnostic, and therapeutic strategies for IUGR, perhaps affecting the health of future generations. References 1. Hooken-Koelega ACS. Intrauterine growth retardation. Int Growth Monitor. 2001;11:2-8. 2. Lin CC, Su SJ, River LP. Comparison of associated high-risk factors and perinatal outcome between symmetric and asymmetric fetal intrauterine growth retardation. Am J Obstet Gynecol. 1991;164:1535-1541. 3. Menendez C, Ordi J, Ismail MR, et al. The impact of placental malaria on gestational age and birth weight. J Infect Dis. 2000;181:1740-1745. 4. Crosby WM. Studies in fetal malnutrition. Am J Dis Child. 1991;145:871-876. 5. Resnik R. Intrauterine growth restriction. Obstet Gynecol. 2002;99:490-496. 6. Snijders RJM, Sherrod C, Gosden CM, Nicolaides KH. Fetal growth retardation: associated malformations and chromosomal abnormalities. Am J Obstet Gynecol. 1993;168:547555. 7. Khoury MJ, Erickson D, Cordero JE, McCarthy BJ. Congenital malformations and intrauterine growth retardation: a population study. Pediatrics. 1988;82:83-90. 8. Lin C-C, Santolaya-Forgas J. Current concepts of fetal growth restriction: part I. Causes, classification, and pathophysiology. Obstet Gynecol. 1998;92:1044-1055. 9. Martinelli P, Grandone E, Colaizzo D, et al. Familial thrombophilia and the occurrence of fetal growth restriction. Haematologica. 2001;86:428-431. 10. Heilmann L, von Tempelhoff GF, Pollow K. Antiphospholipid syndrome in obstetrics. Clin Appl Thromb Hemost. 2003;9:143-150. 11. Barker DJP. Mothers, Babies and Disease in Later Life. London, UK: B.M.J. Publishing; 1994. 12. Roseboom TJ, van der Meulen JHP, Ravelli ACJ, Osmond C, Barker DJB, Bleker O. Effects of prenatal exposure to the Dutch famine on adult disease later in life: an overview. Mol Cell Endocrinol. 2001;185:93-98. 13. Prentice AM, Cole TJ. Seasonal changes in growth and energy status in the Third World. Proc Nutr Soc. 1994;53:509-519. 14. Schwart ID. Failure to thrive: an old nemesis in the new millenium. Pediatr Rev. 2000;21:257-264. 15. Haworth JC, Ellestad-Sayed JJ, King J, Dilling LA. Fetal growth retardation in cigarette-smoking mothers is not due to decreased maternal food intake. Am J Obstet Gynecol. 1980;137:719-723. 16. Meyer MB. How does maternal smoking affect birth weight and maternal weight gain? Evidence from the Ontario Perinatal Mortality Study. Am J Obstet Gynecol. 1978;131:888-893. 17. Cliver SP, Goldenberg RL, Cutter GR, Hoffman HJ, Davis RO, Nelson KG. The effect of cigarette smoking on neonatal anthropometric measurements. Obstet Gynecol. 1995;85:625-630. 18. Andres RL, Day MC. Perinatal complications associated with maternal tobacco use. Semin Neonatol. 2000;5:231241. 19. Lee PA, Chernausek SD, Hokken-Koelega ACS, Czernichow P. International Small for Gestational Age Advisory Board Consensus Development Conference statement: management of short children born small for gestational age. Pediatr. 2003;111:1253-1261. 20. Lim KH, Zhou Y, Janatopour M, et al. Human cytotrophoblast differentiation/invasion is abnormal in preeclampsia. Am J Pathol. 1991;151:1809-1818. 21. Khong TY, De Wolf F, Robertson WB, Brosens I. Inadequate maternal vascular response to placentation in pregnancies complicated by preeclampsia and by small- 9 Brodsky, Christou 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 10 for-gestational-age infants. Br J Obstet Gynaecol. 1986;93:1049-1059. Kingdom J, Huppertz B, Seaward G, Kaufmann P. Development of the placental villous tree and its consequences for fetal growth. Obstet Gynecol. 2000;92:35-43. Smith SC, Baker PN, Symonds EM. Increased placental apoptosis in intrauterine growth restriction. Am J Obstet Gynecol. 1997;177:1395-1401. Axt R, Kordina AC, Meyberg R, Reitnauer K, Mink D, Schmidt W. Immunohistochemical evaluation of apoptosis in placentae from normal and intrauterine growth-restricted pregnancies. Clin Exp Obstet Gynecol. 1999;26:195-198. Baker J, Liu JP, Robertson EJ, Efstratiadis A. Role of insulinlike growth factors in embryonic and postnatal growth. Cell. 1993;75:73-82. Fowden AL. The insulin-like growth factors and feto-placental growth. Placenta. 2003;24:803-812. Wang HS, Lim J, English J, Irvine L, Chard T. The concentration of insulin-like growth factor-I and insulin-like growth factor binding protein-1 in human umbilicial cord serum at delivery: relation to fetal weight. J Endocrinol. 1991;129:459-464. Giudice LC, de Zegher F, Gargosky SE, Dsupin BA, de las Fuentes L, Crystal RA. Insulin-like growth factors and their binding proteins in the term and preterm human fetus and neonate with normal and extremes of intrauterine growth. J Clin Endocrinol Metab. 1995;80:1549-1555. Giudice LC, Martina NA, Crystal RA, Taguke S, Druzin M. Insulin-like growth factor binding protein-1 at the maternal-fetal interface and insulin-like growth factor-I, insulinlike growth factor II, and insulin-like growth factor binding protein-1 in the circulation of women in severe preeclampsia. Am J Obstet Gynecol. 1997;176:751-758. Woods KA, Camacho-Hubner C, Savage MO, Adrian JL. Brief report: intrauterine growth retardation and postnatal growth failure associated with deletion of the insulin-like growth factor I gene. N Engl J Med. 1996;335:1363-1367. Abuzzahab MJ, Schneider A, Goddard A, et al. IGF-I receptor mutations resulting in intrauterine and postnatal growth retardation. N Engl J Med. 2003;349:2211-2222. Crossey PA, Pillai CC, Miell JP. Altered placental development and intrauterine growth restriction in IGF binding protein-1 transgenic mice. J Clin Invest. 2002;110:411-418. Liu JP, Baker J, Perkins AS, Robertson EJ, Efstratiadis A. Mice carrying null mutations of the genes encoding insulinlike growth factor I (IGF-1) and type I IGF receptor. Cell. 1993;75:59-72. Gluckman PD. The endocrine regulation of fetal growth in late gestation-the role of insulin like growth factors. J Clin Endocrinol Metab. 1995;80:1047-1050. Hay WW Jr, Catz CS, Grave GD, Yaffe SJ. Workshop summary: fetal growth—its regulation and disorders. Pediatrics. 1997;99:585-591. Evain-Brion D. Maternal endocrine adaptations to placental hormones in humans. Acta Paediatr Suppl. 1999;88:1216. Robinson JS, Moore VM, Owens JA, McMillen IC. Origins of fetal growth restriction. Eur J Obstet Gynecol. 2000;92:1319. Janatpour MJ, Utset MF, Cross JC, et al. A repertoire of differentially expressed transcription factors that offers insight into mechanisms of human cytotrophoblast differentiation. Dev Genet. 1999;25:146-157. Anson-Cartwright L, Dawson K, Holmyard D, Fisher SJ, Lazzarini RA, Cross JC. The glial cells missing-1 protein is essential for branching morphogenesis in the chorioallantoic placenta. Nat Genet. 2000;25:311-314. Schreiber J, Riethmacher-Sonnenberg E, Riethmacher D, et al. Placental failure in mice lacking the mammalian homolog of glial cells missing, GCMa. Mol Cell Biol. 2000;20:2466-2474. 41. Christou H, Serdy S, Mantzoros CS. Leptin in relation to growth and developmental processes in the fetus. Semin Reprod Med. 2002;20:123-130. 42. Jansson T, Powell TL. Placental nutrient transfer and fetal growth. Nutrition. 2000;16:500-502. 43. Malandro MS, Beveridge MJ, Kihlberg MS, Novak DA. Effect of low-protein diet-induced intrauterine growth retardation on rate placental amino acid transport. Am J Phsyiol. 1996;271:C295-303. 44. Gilbert WM, Danielsen B. Pregnancy outcomes associated with intrauterine growth restriction. Am J Obstet Gynecol. 2003;188:1596-1601. 45. Gulmezoglu M, de Onis M, Villar J. Effectiveness of interventions to prevent or treat impaired fetal growth. Obstet Gynecol Surv. 1997;52:139-148. 46. Hay WW, Thureen PJ, Anderson MS. Intrauterine growth restriction. NeoReviews. 2001;2:e129-137. 47. Group EC. Randomized trial of low-dose aspirin for the prevention of maternal and fetal complications in high-risk pregnant women. Br J Obstet Gynaecol. 1996;103:39-47. 48. Nicolaides KH, Campbell S, Bradley RJ, Bilardo CM, Soothill PW, Gibb D. Maternal oxygen therapy for intrauterine growth retardation. Lancet. 1987;1:942-945. 49. Bekedam DJ, Mulder EJ, Snijders RJ, Visser GH. The effects of maternal hyperoxia on fetal breathing movements, body movements and heart rate variation in growth retarded fetuses. Early Hum Dev. 1991;27:223-232. 50. Arduini D, Rizzo G, Mancuso S, Romanini C. Short-term effects of maternal oxygen administration on blood flow velocity waveforms in health and growth-retarded fetuses. Am J Obstet Gynecol. 1988;159:1077-1080. 51. Say L, Gulmezoglu AM, Hofmeyr GJ. Maternal oxygen administration for suspected impaired fetal growth. Cochrane Database Syst Rev. 2003;1:CD000137. 52. Ribbert LSM, van Lingen RA, Visser GHA. Continuous maternal hyperoxygenation in the treatment of early fetal growth retardation. Ultrasound Obstet Gynecol. 1991;1:331335. 53. Gulmezoglu AM, Hofmeyr GJ. Bed rest in hospital for suspected impaired fetal growth. Cochrane Database Syst Rev. 2000;2:CD000034. 54. Seeds JW, Peng T. Impaired growth and risk of fetal death: is the tenth percentile the appropriate standard? Am J Obstet Gynecol. 1998;178:658-69. 55. Elimian A, Verma U, Canterino J, Shah J, Visintainer P, Tejani N. Effectiveness of antental steroids in obstetrics subgroups. Obstet Gynecol. 1999;93:174-179. 56. Schaap AH, Wolf H, Bruinse HW, Smolders-De Haas H, Van Ertbruggen I, Treffers EE. Effects of antenatal corticosteroid administration on mortality and long-term morbidity in early preterm growth restricted infants. Obstet Gynecol. 2001;97:954-960. 57. Dubinsky T, Lau M, Powell F, et al. Predicting poor neonatal outcome: a comparative study of noninvasive antenatal testing methods. Am J Roentgenol. 1997;168:827-831. 58. Devoe LD, Jones CR. Nonstress test: evidence-based use in high-risk pregnancy. Clin Obstet Gynecol. 2002;45:986-992. 59. Manning FA, Harman CR, Morrison I, Menticoglou SM, Lange IR, Johnson JM. Fetal assessment based on fetal biophysical profile scoring. IV: an analysis of perinatal morbidity and mortality. Am J Obstet Gynecol. 1990;162:703709. 60. Soothill PW, Ajayi RA, Campbell S, Nicolaides KH. Prediction of morbidity in small and normally grown fetuses by fetal heart rate variability, biophysical profile score and umbilical artery Doppler studies. Br J Obstet Gynaecol. 1993;100:742-745. 61. Ferrazzi E, Vegni C, Bellotti M, Borboni A, Della Peruta S, Barbera A. Role of umbilical Doppler velocimetry in the biophysical assessment of the growth-retarded fetus: answers from neonatal morbidity and mortality. J Ultrasound Med. 1991;10:309-315. Journal of Intensive Care Medicine 18(X); 2004 Intrauterine Growth Restriction 62. Karsdorp VH, van Vugt JM, van Geijn HP, et al. Clinical significance of absent or reversed end diastolic velocity waveforms in umbilical artery. Lancet. 1994;344:1664-1668. 63. Neilson JP, Alfirevic Z. Doppler ultrasound for fetal assessment in high risk pregnancies. Cochrane Database Syst Rev. 2000;2:CD000073. 64. Divon MY, Ferber A. Doppler evaluation of the fetus. Clin Obstet Gynecol. 2002;45:1015-1025. 65. Laurini R, Laurin J, Marsal K. Placental histology and fetal blood flow in intrauterine growth retardation. Acta Obstet Gynecol Scand. 1994;73:529-534. 66. Ferrazzo E, Bulfamante G, Mezzopane R, Barbera A, Ghidini A, Pardi G. Uterine Doppler velocimetry and placental hypoxic-ischemic lesion in pregnancies with fetal intrauterine growth restriction. Placenta. 1999;20:389-394. 67. Berg AT. Childhood neurological morbidity and its association with gestational age, intrauterine growth retardation and perinatal stress. Paediatr Perinat Epidemiol. 1988;2:229-238. 68. Ley D, Laurin J, Bjerre I, Marsal K. Abnormal fetal aortic velocity waveform and minor neurological dysfunction at 7 years of old. Ultrasound Obstet Gynecol. 1996;8:152-159. 69. Bekedam DJ, Visser GH, van der Zee AG, Snijders RJ, Poelmann-Weesjes G. Abnormal velocity waveforms of the umbilical artery in growth retarded fetuses: relationship to antepartum late heart rate decelerations and outcome. Early Hum Dev. 1990;24:79-89. 70. Montenegro N, Santos F, Tavares E, Matias A, Barros H, Leite LP. Outcome of 88 pregnancies with absent or reversed end-diastolic blood flow in the umbilical arteries. Eur J Obstet Gynecol Reprod Biol. 1998;79:43-46. 71. McParland P, Steel S, Pearce JM. The clinical implications of absent or reduced end-diastolic frequencies in umbilical artery flow velocity waveforms. Eur J Obstet Gynecol Reprod Biol. 1990;37:15-23. 72. Ott WJ. Diagnosis of intrauterine growth restriction: comparison of ultrasound parameters. Am J Perinatol. 2000;19:133-137. 73. Boito S, Struijk PC, Ursem NT, Stijnen T, Wladimiroff JW. Umbilical venous volume flow in the normally developing and growth-restricted human fetus. Ultrasound Obstet Gynecol. 2002;19:344-349. 74. Hecher K, Campbell S, Doyle P, Harrington K, Nicolaides K. Assessment of fetal compromise by Doppler ultrasound investigation of the fetal circulation: arterial, intracardiac and venous blood flow velocity studies. Circulation. 1995;91:129-138. 75. Pardi G, Cetin I, Marconi AM, et al. Diagnostic value of blood sampling in fetuses with growth retardation. N Engl J Med. 1993;328:728-729. 76. Chan FY, Pun TC, Lam P, Lam C, Lee CP, Lam YH. Fetal cerebral Doppler studies as a predictor of perinatal outcome and subsequent neurologic handicap. Obstet Gynecol. 1996;87:981-988. 77. Scherjon SA, Oosting H, Smolders-DeHaas H, Zondervan HA, Kok JH. Neurodevelopmental outcome at three years of age after fetal “brain-sparing.” Early Hum Dev. 1998;52:67-79. 78. Bahado-Singh RO, Kovanci E, Jeffres A, et al. The Doppler cerebroplacental ratio and perinatal outcome in intrauterine growth restriction. Am J Obstet Gynecol. 1999;180:750756. 79. Einspieler C, Prechtl HF, Ferrari F, Cioni G, Bos AF. The qualitative assessment of general movements in preterm, term, and young infants: review of the methodology. Early Hum Dev. 1997;50:47-60. 80. Prechtl HFR. Qualitative changes of spontaneous movements in fetus and preterm infant are a marker of neurological dysfunction. Early Hum Dev. 1990;23:151-158. 81. Bos AF, Einspieler C, Prechtl HFR. Intrauterine growth retardation, general movements, and neurodevelopmental outcome: a review. Dev Med Child Neurol. 2001;43:61-68. Journal of Intensive Care Medicine 18(X); 2004 82. Bos AF, van Loon AJ, Martjin A, van Asperen RM, Okken A, Prechtl HFR. Spontaneous motility in preterm, small for gestational age infants. II: qualitative aspects. Early Hum Dev. 1997;50:131-147. 83. Harman CR, Baschat AA. Comprehensive assessment of fetal wellbeing: which Doppler tests should be performed? Curr Opin Obstet Gynecol. 2003;15:147-157. 84. Williams KP, Farquaharson DF, Bebbington M, et al. Screening for fetal well-being in a high-risk pregnant population comparing the nonstress test with umbilical artery Doppler velocimetry: a randomized controlled clinical trial. Am J Obstet Gynecol. 2003;188:1366-1371. 85. Owen P, Harrold AJ, Farrell T. Fetal size and growth velocity in the prediction of intrapartum caesarean section for fetal distress. Br J Obstet Gynaecol. 1997;104:445-449. 86. Ounsted M, Moar V, Scott WA. Perinatal morbidity and mortality in small-for-date babies: the relative importance of some maternal factors. Early Hum Dev. 1981;5:367-375. 87. Witter FR. Perinatal mortality and intrauterine growth retardation. Curr Opin Obstet Gynecol. 1993;5:56-59. 88. Piper JM, Xenakis EM, McFarland M, Elliott BD, Berkus MD, Langer O. Do growth-retarded premature infants have different rates of perinatal morbidity and mortality than appropriately grown premature infants? Obstet Gynecol. 1996;87:169-174. 89. Shankar M, Navti O, Amu O, Konje JC. Assessment of stillbirth risk and associated risk factors in a tertiary hospital. J Obstet Gynecol. 2002;22:34-38. 90. Bernstein IM, Horber JD, Badger GJ, Ohlsson A, Golan A. Morbidity and mortality among very-low-birth-weight neonates with intrauterine growth restriction. The Vermont Oxford Network. Am J Obstet Gynecol. 2000;182:198-206. 91. Kramer MS, Olivier M, McLean FH, Willis DM, Usher RH. Impact of intrauterine growth retardation and body proportionality on fetal and neonatal outcome. Pediatrics. 1990;86:707-713. 92. Low JA, Austin RW, Pancham SR. Fetal asphyxia during the antepartum period in intrauterine growth retarded infants. Am J Obstet Gynecol. 1972;113:351-357. 93. Dijxhoorn MJ, Visser GH, Touwen BC, Huisjes HJ. Apgar score, meconium and acidaemia at birth in small-for-gestational age infants born at term, and their relation to neonatal neurological morbidity. Br J Obstet Gynaecol. 1987;94:873-879. 94. Doctor BA, O’Riordan MA, Kirchner HL, Shah D, Hack M. Perinatal correlates and neonatal outcomes of small for gestational age infants born at term gestation. Am J Obstet Gynecol. 2001;185:652-659. 95. Beiner ME, Simchen MJ, Sivan E, Chetrit A, Kuint J, Schiff E. Risk factors for neonatal thrombocytopenia in preterm infants. Am J Perinatol. 2003;20:49-54. 96. Ott WJ. Intrauterine growth retardation and preterm delivery. Am J Obstet Gynecol. 1993;168:1710-1715. 97. Bardin C, Zelkowitz P, Papageorgiou A. Outcome of smallfor-gestational age and appropriate-for-gestational age infants born before 27 weeks of gestation. Pediatrics. 1997;100:E4. 98. Gortner L, Wauer RR, Stock GJ, et al. Neonatal outcome in small for gestational age infants: do they really do better? J Perinat Med. 1999;27:484-489. 99. Simchen MJ, Beiner ME, Strauss-Liviathan N, et al. Neonatal outcome in growth-restricted versus appropriately grown preterm infants. Am J Perinatol. 2000;17:187-192. 100. Lal MK, Manktelow BN, Draper ES, Field DJ. Chronic lung disease of prematurity and intrauterine growth retardation: a population-based study. Pediatrics. 2003;111:483-487. 101. Zaw W, Gagnon R, da Sliva O. The risks of adverse neonatal outcome among preterm small for gestational age infants according to neonatal versus fetal growth standards. Pediatrics. 2003;111:1273-1277. 102. Tyson JE, Kennedy K, Broyles S, Rosenfeld CR. The small for gestational age infant: assessment or delayed pul- 11 Brodsky, Christou 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 12 monary maturation? Increased or decreased survival? Pediatrics. 1995;95:534-538. Friedman SA, Schiff E, Kao I, Sibai BM. Neonatal outcome after preterm delivery for preeclampsia. Am J Obstet Gynecol. 1995;172:1785-1788. Bhatt AB, Tank PD, Barmade KB, Damania KR. Abnormal Doppler flow velocimetry in the growth restricted foetus as a predictor for necrotising enterocolitis. J Postgrad Med. 2002;48:182-185. Soregaroli M, Bonera R, Danti L, et al. Prognostic role of umbilical artery Doppler velocimetry in growth-restricted fetuses. J Matern Fetal Neonatal Med. 2002;11:199-203. Thorpe-Beeston JG, Nicolaides KH, Snijders RJ, Felton CV, McGregor AM. Thyroid function in small for gestational age fetuses. Obstet Gynecol. 1991;77:701-706. Nieto-Diaz A, Villar J, Matorras-Weinig R, Valenzuela-Ruiz P. Intrauterine growth retardation at term: association between anthropometric and endocrine parameters. Acta Obstet Gynecol Scand. 1996;75:127-131. Hay WWJ, Lucas A, Heird WC, et al. Workshop summary: nutrition of the extremely low birth weight infant. Pediatrics. 1999;104:1360-1368. Martin CR, Van Marter LJ, Allred EN, Leviton A. Growthrestricted premature infants are at increased risk for low thyroxine. Early Hum Dev. 2001;64:119-128. Dashe JS, McIntire DD, Lucas MJ, Leveno KJ. Effects of symmetric and asymmetric fetal growth on pregnancy outcomes. Obstet Gynecol. 2000;96:321-327. Williams MC, Wyble LE, O’Brien WF, Nelson RM, Schwenke JR, Casanova C. Persistent pulmonary hypertension of the neonate and asymmetric growth restriction. Obstet Gynecol. 1998;91:336-341. Merlet-Benichou C, Gilbert T, Muffat-Joly M, LelievrePegorier M, Leroy B. Intrauterine growth retardation leads to a permanent nephron deficit in the rat. Pediatr Nephrol. 1994;8:175-180. Konje JC, Bell SC, Morton JJ, de Chazal R, Taylor DJ. Human fetal kidney morphometry during gestation and the relationship between weight, kidney morphometry and plasma renin concentration at birth. Clin Sci (Lond). 1996;91:169-175. Bassan H, Trejo LL, Kariv N, et al. Experimental intrauterine growth retardation alters renal development. Pediatr Nephrol. 2000;15:192-195. Bauer R, Walter B, Ihring W, Kluge H, Lampe V, Zwiener U. Altered renal function in growth-restricted newborn piglets. Pediatr Nephrol. 2000;14:735-739. Mackenzie HS, Lawler EV, Brenner BM. Congenital oligonephropathy: the fetal flaw in essential hypertension? Kidney Int Suppl. 1996;55:S30-34. Langely-Evans SC, Welham SJ, Jackson AA. Fetal exposure to a maternal low protein diet impairs nephrogenesis and promotes hypertension in the rat. Life Sci. 1999;64:965-974. Vehaskari VM, Aviles DH, Manning J. Prenatal programming of adult hypertension in the rat. Kidney Int. 2001;59:238-245. Karlberg J, Jalil F, Lam B, Low L, Yeung CY. Linear growth retardation in relation to the three phases of growth. Eur J Clin Nutr. 1994;48:S25-S44. Leger J, Limoni C, Czernichow P. Prediction of the outcome of growth at 2 years of age in neonates with intra-uterine growth retardation. Early Hum Dev. 1997;48:211-223. Steward DK. Biological vulnerability in infants with failure to thrive: the association with birthweight. Child Care Health Dev. 2001;27:555-567. Strauss RS, Dietz WH. Effects of intrauterine growth retardation in premature infants on early childhood growth. J Pediatr. 1997;130:95-102. Boguszewski M, Albertsson-Wikland K, Aronsson S, et al. Growth hormone treatment of short children born smallfor-gestational age: the Nordic Multicenter Trial. Acta Paediatr. 1998;87:257-263. Lee PA, Chernausek SD, Hooken-Koelega ACS, Czernichow P. International SGA Advisory Board 125. 126. 127. 128. 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 144. Consensus Development Conference statement: management of short children born small for gestational age. Pediatrics. 2003;111:1253-1261. Bozzola E, Lauriola S, Messina MF, Bona G, Tinelli C, Tato L. Effect of different growth hormone dosages on the growth velocity in children born small for gestational age. Horm Res. 2004;61:98-102. Van Pareren Y, Mulder P, Houdijk M, Jansen M, Reeser M, Hokken-Koelega A. Adult height after long-term, continuous growth hormone treatment in short children born small for gestational age: results of a randomized, doubleblind, dose response GH trial. J Clin Endocrinol Metab. 2003;88:3584-3590. Carel JC, Chatelain P, Rochiccioli P, Chaussain JL. Improvement in adult height after growth hormone treatment in adolescents with short stature born small for gestational age: results of a randomized controlled study. J Clin Endocrinol Metab. 2003;88:1587-1593. Ranke MB, Lindberg A, Cowell CT, et al. Prediction of response to growth hormone treatment in short children born small for gestational age: analysis of data from KIGS. J Clin Endocrinol Metab. 2003;88:125-131. de Zengher F, Albertsson-Wikland K, Wollmann HA, et al. Growth hormone treatment of short children born small for gestational age: growth responses with continuous and discontinuous regimens over 6 years. J Clin Endocrinol Metab. 2000;85:2816-2821. Sas T, de Waal W, Mulder P, et al. Growth hormone treatment in children with short stature born small for gestational age: 5-year results of a randomized, double-blind, dose response trial. J Clin Endocrinol Metab. 1999;84:30643070. Proceedings of the 13th Novo Nordisk Symposium on Growth Hormone and Endocrinology of the SGA/IUGR Satellite Symposium. Horm Res. 2003;59:1-143. Sommerfelt K, Andersson HW, Sonnander K, et al. Cognitive development of term small for gestational age children at five years of age. Arch Dis Child. 2000;83:25-30. Sommerfelt K, Sonnander K, Skranes J, et al. Neuropsychologic and motor function in small-for-gestation preschoolers. Pediatr Neurol. 2002;26:186-191. Hawdon JM, Hey E, Kolvin I, Fundudis T. Born too small: is outcome still affected? Dev Med Child Neurol. 1990;32:943-953. Pryor J, Silva PA, Brooke M. Growth, development and behaviour in adolescents born small-for-gestational-age. J Paediatr Child Health. 1995;31:403-407. Martyn CN, Gale CR, Sayer AA, Fall C. Growth in utero and cognitive function in adult life: follow up study of people born between 1920 and 1943. BMJ. 1996;312:1393-1396. Paz I, Laor A, Gale R, Harlap S, Stevenson DK, Seidman DS. Term infants with fetal growth restriction are not at increased risk for low intelligence scores at age 17 years. J Pediatr. 2001;138:87-91. O’Keefe MJ, O’Callaghan M, Williams GM, Najman JK, Bor W. Learning, cognitive, and attentional problems in adolescents born small for gestational age. Pediatrics. 2003;112:301-307. Blair E, Stanley F. Intrauterine growth and spastic cerebral palsy. Am J Obstet Gynecol. 1990;162:229-237. Uvebrant P, Hagberg G. Intrauterine growth in children with cerebral palsy. Acta Paediatr. 1992;81:407-412. Topp M, Langhoff-Roos J, Uldall P, Kristensen J. Intrauterine growth and gestational age in preterm infants with cerebral palsy. Early Hum Dev. 1996;44:27-36. Harvey D, Prince J, Burton J, Parkinson C, Campbell S. Abilities of children who were small-for-gestational-age babies. Pediatrics. 1982;69:296-300. Parkinson CE, Wallis S, Harvey D. School achievement and behavior of children who were small-for-dates at birth. Dev Med Child Neurol. 1981;23:41-50. Sung IK, Vohr B, Oh W. Growth and neurodevelopmental outcome of very low birth weight infants with intrauterine growth retardation: comparison with control subjects Journal of Intensive Care Medicine 18(X); 2004 Intrauterine Growth Restriction 145. 146. 147. 148. 149. 150. 151. 152. 153. 154. 155. 156. 157. 158. 159. matched by birth weight and gestational age. J Pediatr. 1993;123:618-624. Wienerroither H, Steinder H, Tomaselli J, Lobendanz M, Thun-Hohenstein L. Intrauterine blood flow and long-term intellectual, neurologic, and social development. Obstet Gynecol. 2001;97:449-453. Vossbeck S, deCamargo OK, Grab D, Bode H, Pohlandt F. Neonatal and neurodevelopmental outcome in infants born before 30 weeks of gestation with absent or reversed end-diastolic flow velocities in the umbilical artery. Eur J Pediatr. 2001;160:128-134. Berg AT. Indices of fetal growth retardation, perinatal hypoxia-related factors and childhood neurological morbidity. Early Hum Dev. 1989;19:271-283. Low JA, Handley-Derry MH, Burke SO, et al. Association of intrauterine fetal growth retardation and learning deficits at age 9 and 11 years. Am J Obstet Gynecol. 1992;167:14991505. Sommerfelt K, Troland K, Ellertsen B, Markestad T. Behavioral problems in low-birthweight preschoolers. Dev Med Child Neurol. 1996;38:927-940. Halsey CL, Collin MF, Anderson CL. Extremely low-birthweight children and their peers: a comparison of schoolage outcomes. Arch Pediatr Adolesc Med. 1996;150:790794. Goldenberg R, Hack M, Grantham-McGregor SM, Schurch B. Report of the IDECG/IUNS Working Group on IUGR effects on neurological, sensory, cognitive, and behavioral function. Eur J Clin Nutr. 1998;52:S100-S101. Buck GM, Cookfair DL, Michalek AM, et al. Intrauterine growth retardation and risk of sudden infant death syndrome (SIDS). Am J Epidemiol. 1989;129:874-884. Wierenga H, Brand R, Geudeke T, van Geijn HP, van der Harten H, Verloove-Vanhorick SP. Prenatal risk factors for cot death in very preterm and small for gestational age infants. Early Hum Dev. 1990;23:15-26. Filiano JJ, Kinney HC. A perspective on neuropathologic findings in victims of the sudden infant death syndrome: the triple-risk model. Biol Neonate. 1994;65:194-197. Harding R, Tester ML, Moss TJ, et al. Effects of intra-uterine growth restriction on the control of breathing and lung development after birth. Clin Exp Pharm Phys. 2000;27:114-119. Rona RJ, Gulliford MC, Chinn S. Effects of prematurity and intrauterine growth on respiratory health and lung function in childhood. BMJ. 1993;306:817-820. Barker DJP, Osmond C. Infant mortality, childhood nutrition, and ischeamic heart disease in England and Wales. Lancet. 1986;1:1077-1081. Barker DJP. The developmental origins of adult disease. Eur J Epidemiol. 2003;18:733-736. Huxley R, Neil A, Collins R. Unravelling the fetal origins hypothesis: is there really an inverse association between birthweight and subsequent blood pressure? Lancet. 2002;360:659-665. Journal of Intensive Care Medicine 18(X); 2004 160. Law CM, Shiell AW. Is blood pressure inversely related to birth weight? The strength of evidence from a systematic review of the literature. J Hypertens. 1996;14:935-941. 161. Hinchliffe SA, Lynch MR, Sargent PH, Howard CV, Van Velzen D. The effect of intrauterine growth retardation on the development of renal nephrons. Br J Obstet Gynaecol. 1992;99:296-301. 162. Manalich R, Reyes L, Herrera M, Melendi C, Fundora I. Relationship between weight at birth and the number and size of renal glomeruli in humans: a histomorphometric study. Kidney Int. 2000;58:770-773. 163. Brenner BM, Chertow GM. Congenital oligonephropathy and the etiology of adult hypertension and progressive renal injury. Am J Kid Dis. 1994;23:171-175. 164. Hales CN, Ozanne SE. For debate: fetal and early postnatal growth restriction lead to diabetes, the metabolic syndrome and renal failure. Diabetologia. 2003;46:1013-1019. 165. Hales CN, Ozanne SE. The dangerous road to catch-up growth. J Physiol. 2003;547:5-10. 166. Ozanne SE, Olsen GS, Hansen LL, et al. Early growth restriction leads to down regulation of protein kinase C zeta and insulin resistance in skeletal muscle. J Endocrinol. 2003;177:235-241. 167. Sparre T, Reusens B, Cherif H, et al. Intrauterine prrogramming of fetal islet gene expression in rats: effects of maternal protein restriction during gestation revealed by proteome analysis. Diabetologia. 2003;46:1497-1511. 168. Eriksson JG, Forsen T, Tuomilehto J, Osmond C, Barker DJ. Early adiposity rebound in childhood and risk of type 2 diabetes in adult life. Diabetologia. 2003;46:190-194. 169. Singhal A, Fewtrell M, Cole TJ, Lucas A. Low nutrient intake and early growth for later insulin resistance in adolescents born preterm. Lancet. 2003;361:1089-1097. 170. Eriksson JG, Lindi V, Uusitupa M, et al. The effects of the Pro12Ala polymorphism of the peroxisome proliferatoractivated receptor-g2 gene on insulin sensitivity and insulin metabolism interact with size at birth. Diabetes. 2002;51:2321-2324. 171. Davis L, Roullet JB, Thornburg KL, Shokry M, Hohimer AR, Giraud GD. Augmentation of coronary conductance in adult sheep made anaemic during fetal life. J Physiol. 2003;547:53-59. 172. Broberg CS, Giraud GD, Schultz JM, Thornburg KL, Hohimer AR, Davis LE. Fetal anemia leads to augmented contractile response to hypoxic stress in adulthood. Am J Physiol. 2003;285:R6490-R6455. 173. Philip AGS. Classification by birthweight and gestational age. NeoReviews. 2003;4:e91-93. 174. Cetin I, Marconi AM, Baggiani AM, et al. In vivo placental transport of glycine and leucine in human pregnancies. Pediatr Res. 1995;37:571-575. 175. Lok F, Owens JA, Mundy L, Robinson JS, Owens PC. Insulin-like growth factor I promotes growth selectively in fetal sheep in late gestation. Am J Physiol. 1996;270:R11481155. 13