Preterm Intraventricular Hemorrhage/Posthemorrhagic Hydrocephalus




Abstract


Germinal matrix hemorrhage–intraventricular hemorrhage (GMH-IVH) is the most common variety of neonatal intracranial hemorrhage and is characteristic of the premature infant. This form of brain injury affects around 25% of all very low birthweight (<1500 g) premature infants, resulting in an increased risk for neurodevelopmental disability. Its occurrence can lead to impact on subsequent brain development because of destruction of the immature germinal cerebral region resulting in loss of progenitor cells, acceleration of white matter injury via pressure and oxidative stress, and direct parenchymal injury. Finally, hydrocephalus can complicate IVH, resulting in a further increased risk of neurodevelopmental disability. In this chapter, the neuropathology, pathogenesis, clinical features, diagnosis, prognosis, and management of IVH, and its complications will be reviewed. The prominent position of this lesion in neonatal medicine has been accompanied by a large increase in work from several disciplines. This chapter attempts to integrate this information in a meaningful way without oversimplifying a clearly complex problem.




Keywords

germinal matrix hemorrhage, intraventricular hemorrhage, neonatal parenchymal infarction, posthemorrhagic hydrocephalus, premature infant cerebral injury

 




Neuropathology


The neuropathology of IVH is best considered in terms of the site of origin (primarily the germinal matrix), the spread of the hemorrhage throughout the ventricular system, the neuropathological consequences of the hemorrhage, and the neuropathological accompaniments not necessarily related directly to the IVH.


The basic lesion in germinal matrix hemorrhage–IVH is bleeding into the subependymal germinal matrix. This region is represented by the ventricular-subventricular zone described in Chapter 6 . Over the final 12 to 16 weeks of gestation, this matrix becomes less and less prominent and is essentially exhausted by term (see later discussion). This region is highly cellular, gelatinous in texture, and as would be expected for a structure with active cellular proliferation, richly vascularized. To understand the nature of IVH, it is useful to review first the arterial and venous supply to the germinal matrix.


Arterial Supply to Subependymal Germinal Matrix


The arterial supply to the subependymal germinal matrix is derived particularly from the anterior cerebral artery (through Heubner artery), the middle cerebral artery (primarily through the deep lateral striate branches but also through penetrating branches from surface meningeal branches), and the internal carotid artery (through the anterior choroidal artery; Fig. 24.5 ). The relative importance of these arteries in the vascular supply to the capillaries of the matrix is not entirely clear; different studies have attributed particular importance to the Heubner artery and to the lateral striate arteries. However, it is likely that the terminal branches of this arterial supply constitute a vascular end zone and thus a vulnerability to ischemic injury.




Figure 24.5


Arterial supply.

Arterial supply to the subependymal germinal matrix at 29 weeks of gestation.

(From Hambleton G, Wigglesworth JS. Origin of intraventricular haemorrhage in the preterm infant. Arch Dis Child . 1976;51:651–659.)


Capillary Network


The rich arterial supply just described feeds an elaborate capillary bed in the germinal matrix. This bed generally is composed of relatively large, irregular endothelial-lined vessels that do not exhibit the characteristics of arterioles or venules and are classified as capillaries or channels, or both. Pape and Wigglesworth characterized the anatomical appearance as “a persisting immature vascular rete in the subependymal matrix which is only remodeled into a definite capillary bed when the germinal matrix disappears.” As term approaches, some of the larger endothelial-lined vessels acquire a collagenous adventitial sheath and can be categorized appropriately as veins, as also described in the matrix of the monkey. The nature of the endothelial-lined vessels in this microvascular bed may be of pathogenetic importance concerning germinal matrix hemorrhage.


These germinal matrix vessels exhibit a variety of unique characteristics that may underlie the fragility and propensity to hemorrhage. These characteristics include exuberant angiogenesis, related to high vascular endothelial growth factor (VEGF) and angiopoietin levels, discontinuous glial endfeet of the blood-brain barrier, relative lack of pericytes, immature basal lamina characteristics, and developmentally regulated expression of vascular wall characteristics, including molecules such as alkaline phosphatase, and high morphometric ratio of diameter to wall thickness.


Venous Drainage of Subependymal Germinal Matrix


The rich microvascular network just described is continuous with a well-developed deep venous system. This venous drainage eventually terminates in the great cerebral vein of Galen ( Fig. 24.6 ). In addition to the matrix region, this venous system drains blood from the cerebral white matter, choroid plexus, striatum, and thalamus through the medullary, choroidal, thalamostriate, and terminal veins. Indeed, the terminal vein, which runs essentially within the germinal matrix, is the principal terminus of the medullary, choroidal, and thalamostriate veins. The latter three vessels course primarily anteriorly to a point of confluence at the level of the head of the caudate nucleus to form the terminal veins, which empty into the internal cerebral vein that courses directly posteriorly to join the vein of Galen. Thus at the usual site of germinal matrix hemorrhage, the direction of blood flow changes in a peculiar U-turn. This feature may have pathogenetic implications (see later section). This venous anatomy is also relevant to the occurrence of periventricular hemorrhagic infarction (see later discussion).




Figure 24.6


Veins of the Galenic system, midsagittal view.

Note that the medullary, choroidal, and thalamostriate veins come to a point of confluence to form the terminal vein. The terminal vein, which courses through the germinal matrix, empties into the internal cerebral vein, and the major flow of blood changes direction sharply at that junction.


Site of Origin and Spread of Intraventricular Hemorrhage


Site of Origin


The site of origin of IVH characteristically is the subependymal germinal matrix ( Fig. 24.7 ). This cellular region immediately ventrolateral to the lateral ventricle serves as the source of cerebral excitatory neuronal precursors between approximately 10 to 20 weeks of gestation and in the second half of gestation provides neuroglial precursors that become cerebral oligodendroglia and astrocytes and late migrating GABAergic neurons destined for the cerebral cortex and, especially, the thalamus (see Unit I). Indeed, elegant studies of Del Bigio showed exuberant proliferation of precursor cells in the germinal matrix until 28 weeks of gestation, with a rapid decline thereafter. For reasons discussed earlier, the many thin-walled vessels in the matrix are a ready source of bleeding. The matrix undergoes progressive decrease in size, from a width of 2.5 mm at 23 to 24 weeks, to 1.4 mm at 32 weeks, to nearly complete involution by approximately 36 weeks. The matrix from 28 to 32 weeks is most prominent in the thalamostriate groove at the level of the head of the caudate nucleus at the site of or slightly posterior to the foramen of Monro, and this site is the most common for germinal matrix hemorrhage. Before 28 weeks, hemorrhage in persisting matrix over the body of the caudate nucleus may also be found. Hemorrhage from choroid plexus occurs in nearly 50% of infants with germinal matrix hemorrhage and IVH, and in more mature infants especially, it may be the major site of origin of IVH (see Chapters 20 to 22 ).




Figure 24.7


Germinal matrix–intraventricular hemorrhage.

Coronal sections of cerebrum. (A) Germinal matrix hemorrhage (arrowheads) at the level of the head of the caudate nucleus and foramen of Monro (see probe), with rupture into the lateral ventricles. (B) Massive intraventricular hemorrhage. Obstruction at the foramen of Monro has caused severe, unilateral ventricular dilation.


The vascular site of origin of germinal matrix hemorrhage within the microcirculation of this region appears most commonly to be the prominent endothelial-lined vessels described earlier, not clearly arterial or venous. Particular importance for vessels in free communication with the venous circulation (e.g., capillary-venule junction or small venules) is suggested by the emergence of solution into germinal matrix hemorrhage from postmortem injection into the jugular veins but not from injection into the carotid artery. Histochemical studies of germinal matrix vessels at the site of hemorrhage also are consistent with an origin at the capillary-venule or small venule level. Multiple microcirculatory sites involving small vessels lined only by endothelium may be involved, depending on the clinical circumstances.


Spread of Intraventricular Hemorrhage


In the approximately 80% of cases with germinal matrix hemorrhage in which blood enters the lateral ventricles, spread occurs throughout the ventricular system ( Fig. 24.8 ). Blood proceeds through the foramina of Magendie and Luschka and tends to collect in the basilar cisterns in the posterior fossa; with substantial collections, the blood may incite an obliterative arachnoiditis over days to weeks with obstruction to cerebrospinal fluid (CSF) flow. Other sites at which particulate blood clot may lead to impaired CSF dynamics are the aqueduct of Sylvius and the arachnoid villi (see later discussion of hydrocephalus).




Figure 24.8


Spread of intraventricular hemorrhage.

(A) Coronal and (B) sagittal views. In A, note blood in the lateral ventricles, aqueduct of Sylvius, the fourth ventricle, and the subarachnoid space around the cerebellum and lower brain stem. In B, note blood throughout the ventricular system (the numbers 1 to 4 refer to lateral ventricle, third ventricle, aqueduct, and fourth ventricle, respectively).


Neuropathological Consequences of Intraventricular Hemorrhage


Several neuropathological states occur as apparent consequences of IVH, including germinal matrix destruction, cerebral white matter injury/dysmaturation, cerebral gray matter dysmaturation, cerebellar dysmaturation, periventricular hemorrhagic infarction, and posthemorrhagic hydrocephalus.


Germinal Matrix Destruction


Destruction of germinal matrix and, importantly, its precursor cells for glia, especially oligodendroglial precursor cells (OPCs) and late migrating GABAergic neurons, is a consistent and expected feature of germinal matrix hemorrhage (see Fig. 24.9 ). The hematoma is frequently replaced by a cyst, the walls of which include hemosiderin-laden macrophages and reactive astrocytes. The destruction of glial precursor cells may have a deleterious influence on subsequent brain development, as outlined next (see Fig. 24.9 ).




Figure 24.9


Neuropathologic consequences of germinal matrix–intraventricular hemorrhage on cerebral white and gray matter, and cerebellum.


Cerebral White Matter Injury/Dysmaturation


As described earlier and in Unit I, the germinal matrix (ganglionic eminence during the developmental period of major occurrence of GMH-IVH; i.e., 24 to 32 weeks’ gestation) is a principal source of proliferation of OPCs, which later in the third trimester migrate into the cerebral white matter, differentiate, and after term equivalency produce cerebral myelin. Loss of these myelin-producing cells could lead to impaired cerebral development. Importantly, studies of postmortem human brains with GMH-IVH, as well as experimental models of GMH, have shown impairment of proliferation of OPCs and their subsequent migration and differentiation. Experimental studies suggest that these deleterious effects on OPCs are mediated by blood products, inflammatory compounds, and microglia. Indeed microglial activation in the germinal matrix and periventricular white matter has been shown in the postmortem human brain with GMH ± IVH. The role of microglia in the mediation of cerebral white matter injury/dysmaturation is discussed in more detail in Chapter 15 concerning the pathophysiology of periventricular leukomalacia.


A related possibility for a deleterious effect of GMH-IVH on cerebral white matter involves free-radical mediated effects on differentiating oligodendrocytes (OLs) and perhaps also on rapidly growing axons in the cerebral white matter (see Chapter 15 ), related in part to the release of nonheme iron from the hemorrhage.


Cerebral Gray Matter Dysmaturation


A deleterious effect of mild GMH-IVH on cerebral cortical and thalamic volumetric development is suggested by recent magnetic resonance imaging (MRI) studies (see later). However, data are limited, and careful neuropathological analysis is lacking. If initial findings are corroborated, a role for germinal matrix destruction should be considered (see Fig. 24.9 ). As noted earlier, during the developmental period of the peak occurrence of GMH-IVH, the germinal matrix contributes to the generation and later migration of GABAergic neurons for cerebral cortex but especially also for association nuclei in the thalamus, both critical for high-level cognitive functioning.


Cerebellar Dysmaturation


Cerebellar dysmaturation, principally in the form of diminished cerebellar volumetric growth, in the absence of overt parenchymal destructive disease, is the most common cerebellar abnormality of the premature infant. Although multiple pathogenetic factors likely operate, strong evidence supports an important role for IVH and associated extraaxial blood.


The possibility that the cerebellar underdevelopment in premature infants may be related to adverse effects of blood products has been raised principally by the observations of Messerschmidt and co-workers, who have described the severe end of the spectrum of the acquired cerebellar growth failure. In their series of 35 infants (mean gestational age, 27 weeks; mean birthweight, 900 g), after an initially normal cerebellar ultrasonographic examination in the first week of life, subsequent ultrasound and then MRI scans identified a gradual deficit in volume, without any apparent injury pattern, over the ensuing weeks. The pons and medulla also were found to be small subsequently. Using MRI sequences optimal for detection of hemosiderin, they identified infratentorial hemosiderin deposition in 70% of infants. The deposition was particularly prominent on the cerebellar surface but was also noted on the surface of the brain stem and in the fourth ventricular region ( Fig. 24.10 ). Hemosiderin in the posterior fossa conveyed a sensitivity of 0.70 (95% confidence interval [CI], 0.48 to 0.86) and a specificity of 0.95 (CI, 0.84 to 0.99), with a positive predictive value of 0.88 and a negative predictive value of 0.87, for cerebellar underdevelopment. Nearly all infants had experienced IVH, and 69% had posthemorrhagic hydrocephalus. A later study of 172 preterm infants identified a linear relationship between IVH and decreased cerebellar volumes with advancing postnatal age. A recent report of 72 preterm infants confirmed these observations, even in the presence of grade II IVH. Consistent with the possibility of a direct relationship between extraaxial blood and impaired cerebellar development is a recent report that showed by MRI a significant relationship between the presence of extraaxial blood and diminished cerebellar volumetric growth with advancing postnatal age, equivalent to the third trimester. As discussed in Chapter 4 , this developmental period is characterized by maximal proliferative activity in the external granular layer located on the surface of the cerebellum and crucial for cerebellar growth.




Figure 24.10


Cerebellar underdevelopment and hemosiderin deposition by magnetic resonance imaging (MRI) in a newborn at term equivalent age.

Sagittal T2-weighted MRI (A) from a 13-week-old infant born at 26 weeks’ gestation shows small vermis, enlarged fourth ventricle, reduced dimensions of the brain stem, and inclined tentorium; hemosiderin deposition is apparent on the surface of the pons and the lining of the fourth ventricle (black arrows) . Horizontal MRI (B) shows reduced volume of cerebellar hemispheres with hemosiderin deposition in both hemispheres (black arrows) .

(Reproduced with permission from Messerschmidt A, Prayer D, Brugger PC, Boltshauser E, Zoder G, Sterniste W, Pollak A, Weber M, Birnbacher R. Preterm birth and disruptive cerebellar development: assessment of perinatal risk factors. Eur J Paediatr Neurol . 2008;12:455–460.)


Thus the data raise the strong possibility that the key targets for the adverse effects of blood over the surface of the cerebellum of the small premature infant are the granule precursor cells of the external granular layer . The proliferating cells of the external granular layer are located directly at the interface with the subarachnoid space. Impairment of the survival or proliferation, or both, of these cells could result in the cerebellar underdevelopment, as evidenced by MRI. The effect on the external granular layer would result not only in deficient generation of internal granule cells but also in disturbance of the granular excitatory input to Purkinje cells and other cells of the molecular layer. The result would be deficient development of the full spectrum of cerebellar circuitry.


The mechanisms of disturbance to the external granular layer in the context of hemosiderin deposition almost certainly would relate to the generation of free radicals , especially reactive oxygen species ( Fig. 24.11 ). Hemosiderin is derived from blood by the following sequential steps: hemolysis of red blood cells, formation of heme, conversion of heme to free iron (and biliverdin, carbon monoxide) by heme oxygenase, and formation of ferritin and then hemosiderin. Free iron is toxic because it leads to the generation of reactive oxygen species, especially the hydroxyl radical by the Fenton reaction. In one adult study of brain with hemosiderin deposits, free iron was increased 2.5-fold in the cerebellar cortex and 14.5-fold in the medulla. In experimental models, intracortical injections of free iron lead to lipid peroxidation products and epileptogenic necrotic foci. In addition, hemosiderin, although a storage form of iron, may also release iron from its protein matrix. The central nervous system has limited ability to discharge iron, and thus the accumulated iron can produce a chronic deleterious effect. Notably, studies of CSF of infants with posthemorrhagic hydrocephalus show persistence of copious amounts of nonprotein-bound iron for weeks after IVH.




Figure 24.11


Likely mechanisms by which direct adverse effects on the external granule cell layer lead to diminished volumetric development of cerebellum and pontine and olivary nuclei.

(From Volpe JJ. Cerebellum of the premature infant: rapidly developing, vulnerable, clinically important. J Child Neurol . 2009;24:1085–1104.)


Periventricular Hemorrhagic Infarction


Approximately 10% to 15% of VLBW infants with IVH also exhibit a characteristic parenchymal lesion (i.e., a relatively large region of hemorrhagic necrosis in the periventricular white matter), just dorsal and lateral to the external angle of the lateral ventricle ( Fig. 24.12 ). The incidence of the lesion increases with decreasing gestational age, such that in infants of less than 750 g, periventricular hemorrhagic infarction accounts for nearly 15% of all cases with IVH (see later). a


a References .

The distribution of high-grade IVH (grades 3 and 4 IVH) for each gestational age from 1993 to 2013 shows the very high incidence of severe IVH in the most immature infants (see Fig. 24.4 ).


Figure 24.12


Periventricular hemorrhagic infarction with intraventricular hemorrhage; coronal sections of cerebrum.

(A) Early lesion; note evolving hemorrhagic infarction (arrowheads) on the same side as larger intraventricular hemorrhage. (B) More advanced lesion; note hemorrhagic necrosis with liquefaction in periventricular white matter (arrowheads) on the same side as larger intraventricular hemorrhage. The ependymal lining is marked by white arrows.


Large-scale ultrasonographic studies have defined the topographic characteristics of periventricular hemorrhagic infarction . The parenchymal hemorrhagic necrosis is strikingly asymmetrical; in the largest early series reported, 67% of such lesions were exclusively unilateral, and in virtually all the remaining cases, lesions were grossly asymmetrical, although bilateral. Approximately one-half of the lesions were extensive and involved the periventricular white matter from frontal to parieto-occipital regions ( Fig. 24.13 ); the remainder were more localized. Approximately 80% of cases were associated with large IVH. Commonly (and mistakenly), the parenchymal hemorrhagic lesion is described as an extension of IVH. Several neuropathological studies have shown that simple extension of blood into cerebral white matter from the germinal matrix or lateral ventricle does not account for the periventricular hemorrhagic necrosis. In a later ultrasonographic report of 58 infants, findings were similar: the lesion was unilateral in 74%, extensive (involving two or more lobar territories) in 67%, and associated with large IVH in 88%. The lobar distribution indicates that the majority of lesions involved the frontal and parietal regions. Approximately 50% of the cases exhibited a midline shift of cerebral structures, consistent with the severity of the lesions. A more recent report of somewhat more localized lesions showed that a majority of the lesions were predominantly parietal with fewer in the frontal and temporal regions. This lobar predominance has implications for outcome (see later).




Figure 24.13


Periventricular hemorrhagic infarction, neuropathology.

Horizontal section of cerebrum above the level of lateral ventricles from a premature infant who died on the sixth postnatal day, 3 days after severe intraventricular hemorrhage. Hemorrhagic necrosis in left cerebral white matter separated from the brain section during fixation and revealed a shaggy margin of the hemorrhagic infarction. See text for details.


Microscopic study of the periventricular hemorrhagic necrosis just described indicates that the lesion is a hemorrhagic infarction . The careful studies of Gould and co-workers and Takashima and co-workers emphasized that (1) the hemorrhagic component consists usually of perivascular hemorrhages that follow closely the fan-shaped distribution of the medullary veins in periventricular cerebral white matter ( Fig. 24.14A and B ), and (2) the hemorrhagic component tends to be most concentrated near the ventricular angle where these veins become confluent and ultimately join the terminal vein in the subependymal region. Thus the periventricular hemorrhagic necrosis occurring in association with large IVH is, in fact, a venous infarction. The most common neuropathological sequela of periventricular hemorrhagic infarction is a large porencephalic cyst at the site of the lesion, either alone (66%) or in combination with smaller cysts (23%). The large cyst communicates often, although not invariably, with the lateral ventricle.






Figure 24.14


Venous drainage of cerebral white matter in schematic and actual appearances.

(A) Schematic diagram shows that the medullary veins, arranged in a fan-shaped distribution, drain blood from the cerebral white matter into the terminal vein, which courses through the germinal matrix. (B) Postmortem venogram obtained from a human newborn shows the actual appearance of the vessels. (C) Periventricular hemorrhagic infarction: coronal magnetic resonance imaging scan (fast spin-echo image) demonstrating bilateral germinal matrix–intraventricular hemorrhages, with an apparent periventricular hemorrhagic infarction on the side of the larger amount of germinal matrix and intraventricular blood (reader’s right). Note the fan-shaped linear distribution of increased signal in the parenchymal lesion (reader’s right), consistent with a combination of intravascular thrombi and perivascular hemorrhage along the course of the medullary veins. LV , Lateral ventricle.

(B From Takashima S, Mito T, Ando Y. Pathogenesis of periventricular white matter hemorrhages in preterm infants. Brain Dev . 1986;8:25–30. C From Counsell SJ, Maalouf EF, Rutherford MA, Edwards AD. Periventricular haemorrhagic infarct in a preterm neonate. Eur J Paediatr Neurol . 1999;3:25–28.)


Periventricular hemorrhagic infarction is distinguishable neuropathologically from secondary hemorrhage into periventricular leukomalacia, which is the ischemic, usually nonhemorrhagic, and symmetrical lesion of periventricular white matter of the premature infant (see later discussion). Distinction of these two lesions in vivo, however, is sometimes difficult. Indeed, because the pathogeneses of periventricular hemorrhagic infarction and periventricular leukomalacia overlap (see later discussion), it is to be expected that the lesions often coexist, thereby sometimes causing confusion in interpretation of cranial ultrasound scans. In Table 24.2 , the basic features of these two periventricular white matter lesions of the premature infant are compared.



TABLE 24.2

Periventricular White Matter Lesions in the Premature Infant With Intraventricular Hemorrhage


























LESION
PERIVENTRICULAR HEMORRHAGIC INFARCTION PERIVENTRICULAR LEUKOMALACIA
Likely site of circulatory disturbance Venous Arterial
Grossly hemorrhagic Invariable Uncommon
Markedly asymmetrical Nearly invariable Uncommon
Evolution Single large cyst Multiple small cysts


The pathogenesis of periventricular hemorrhagic infarction appears to be related causally to the GMH-IVH. A direct relation to the latter lesion seems likely on the basis of three fundamental findings. First, 80% to 90% of the reported parenchymal lesions are observed in association with large (and almost invariably) asymmetrical GMH-IVH. Second, the parenchymal lesions invariably occurred on the same side as the larger amount of germinal matrix and intraventricular blood ( Table 24.3 ). Third, in some cases, the lesions were shown to develop and progress after the occurrence of the GMH-IVH. More than one-half of the lesions were detected after the second postnatal day, when approximately 75% of cases of IVH have already occurred (see the section on diagnosis). The association of large asymmetrical GMH-IVH with progression to ipsilateral periventricular hemorrhagic infarction has been confirmed. These data suggest that the IVH or its associated germinal matrix hemorrhage leads to obstruction of the terminal veins and thus impaired blood flow in the medullary veins with the occurrence of hemorrhagic venous infarction. A similar conclusion was suggested from a neuropathological study. The timing of this progression to infarction is often very rapid because, in most cases, the severe IVH and the periventricular hemorrhagic infarction are detected simultaneously.



TABLE 24.3

Laterality of Apparent Periventricular Hemorrhagic Infarction and Concurrent Asymmetrical Intraventricular Hemorrhage
















SEVERITY OF INTRAVENTRICULAR HEMORRHAGE PERIVENTRICULAR HEMORRHAGIC INFARCTION HOMOLATERAL PERIVENTRICULAR HEMORRHAGIC INFARCTION CONTRALATERAL
Grade III 47 0
Grades I–II 5 4

Data from Guzzetta F, Shackelford GD, Volpe S, Perlman JM, Volpe JJ. Periventricular intraparenchymal echodensities in the premature newborn: critical determinant of neurologic outcome. Pediatrics. 1986;78:995–1006.


This pathogenetic formulation received strong support from Doppler determinations of blood flow velocity in the terminal vein during the evolution of the infarction in the living premature infant; obstruction of flow in the terminal vein by the ipsilateral GMH-IVH was shown clearly. Moreover, the finding of elevated lactate in structures adjacent to the GMH, in the distribution of tributaries of the terminal vein, further supports the occurrence of ischemia secondary to venous obstruction by the matrix hemorrhage. Finally, an MRI study of acute periventricular hemorrhagic infarction has shown an appearance consistent with a combination of intravascular thrombi and perivascular hemorrhage along the course of the medullary veins within the area of infarction (see Fig. 24.14C ).


The pathogenetic scheme that is considered to account for most examples of periventricular hemorrhagic infarction is shown in Fig. 24.15 . This scheme should be distinguished from that operative for hemorrhagic periventricular leukomalacia ( Fig. 24.16 ), although the lesions could coexist. The frequency of coexistence of the two lesions is not known. In addition, the two pathogenetic schemes could operate in sequence; that is, periventricular leukomalacia could become secondarily hemorrhagic (and perhaps a larger area of injury) when GMH or IVH subsequently causes venous obstruction.




Figure 24.15


Pathogenesis of periventricular hemorrhagic infarction.

The formulation indicates a central role for germinal matrix–intraventricular hemorrhage in causation of the periventricular venous infarction.



Figure 24.16


Pathogenesis of hemorrhagic periventricular leukomalacia.


The deleterious neurological effects of periventricular hemorrhagic infarction may relate not only to destruction of cerebral white matter per se but also to the effects of the white matter injury on cerebral cortical development. Thus careful neuropathological study of cerebral cortical organization in infants who died with major hemorrhagic white matter lesions has shown striking alterations in neuronal axonal and dendritic ramifications in areas overlying the white matter destruction. Moreover, in unpublished work from our group, cerebral cortical gray matter volume was shown by three-dimensional MRI to be reduced at term in premature infants with periventricular hemorrhagic infarction. These abnormalities are postulated to be secondary to disturbances of afferent input to and efferent input from the areas of cortex by disruption of the respective white matter axons. Another potential cause of the cortical abnormalities could be the destruction of subplate neurons by the white matter infarction. These neurons are critical for cerebral cortical organization and are abundant in subcortical white matter in the human premature infant (see Unit 1). Whatever the mechanism, these cerebral cortical abnormalities with periventricular hemorrhagic infarction could be very important in determining subsequent cognitive deficits and seizure disorders.


Hydrocephalus


An additional neuropathological consequence of IVH is progressive posthemorrhagic ventricular dilation (i.e., hydrocephalus). The likelihood and the rapidity of evolution of hydrocephalus after IVH are related directly to the quantity of intraventricular blood. Thus, with large IVH, hydrocephalus may evolve over days ( acute hydrocephalus), and with smaller IVH, the process evolves usually over weeks ( subacute-chronic hydrocephalus; see later discussion).


Acute hydrocephalus is accompanied by particulate blood clot, readily demonstrated in life by ultrasound scan (see later discussion). The particulate clot may impair CSF absorption by obstruction of the arachnoid villi. This mechanism may be particularly likely in the newborn, in whom only microscopic arachnoid villi (and not larger, later appearing arachnoid granulations) are present. The possibility that endogenous fibrinolytic mechanisms mediated by plasminogen activation are deficient in the CSF of the premature infant is suggested by the findings that plasminogen levels are extremely low in the CSF of such infants, whereas in infants with recent IVH, the levels of plasminogen activator inhibitor are relatively high. This combination of findings may limit the infant’s capacity to mediate clot lysis after IVH.


Subacute-chronic hydrocephalus relates most commonly either to an obliterative arachnoiditis in the posterior fossa (which results in either obstruction of fourth ventricular outflow or flow through the tentorial notch) or to aqueductal obstruction by blood clot, disrupted ependyma, and reactive gliosis. The obliterative arachnoiditis is probably most important. Two molecules important in fibroproliferative responses have been shown to be upregulated in infants with posthemorrhagic hydrocephalus. Transforming growth factor-beta 1, derived in this setting from platelets, is a cytokine chemotactic for fibroblasts and important in the upregulation of genes encoding collagen, fibronectin, and other extracellular matrix proteins. Procollagen 1C-peptide, involved in collagen fiber formation and tissue deposition, also has been shown to be elevated in CSF of infants with posthemorrhagic hydrocephalus.


The deleterious effects of hydrocephalus on cerebral white matter are discussed later (see section on progressive posthemorrhagic ventricular dilatation). Prominent affection of white matter axons and microcirculation is emphasized.


Neuropathological Accompaniments of Intraventricular Hemorrhage


Several neuropathological states are common accompaniments of IVH, but, in contrast to the states just described, these are generally not caused by the IVH. The two most common accompaniments are periventricular leukomalacia and selective neuronal necrosis.


Periventricular Leukomalacia


Periventricular leukomalacia , the generally symmetrical, nonhemorrhagic, and apparently ischemic white matter injury of the premature infant (see Chapter 14 , Chapter 15 , Chapter 16 ), was observed to some degree in 75% of one series of infants who died with IVH. The frequent association of classic necrotic/cystic periventricular leukomalacia and IVH also was emphasized in three other neuropathological reports, as well as in two large ultrasonographic studies. Although it has been reported that approximately 25% of examples of periventricular leukomalacia become hemorrhagic, especially when associated coagulopathy is present, this figure includes examples that have been accompanied by large IVH and that probably represent the venous infarction discussed earlier as periventricular hemorrhagic infarction. Takashima and co-workers suggested that the two lesions (i.e., periventricular hemorrhagic infarction and hemorrhagic periventricular leukomalacia) may be distinguishable in part on the basis of topography. Thus hemorrhagic periventricular leukomalacia has a predilection for periventricular arterial border zones, particularly in the region near the trigone of the lateral ventricles. Venous infarction, especially its most hemorrhagic component, is particularly prominent more anteriorly; that is, the lesion radiates from the periventricular region at the site of confluence of the medullary and terminal veins and assumes a roughly triangular, fan-shaped appearance in periventricular white matter.


IVH also may contribute to the occurrence of periventricular leukomalacia . The possibility of periventricular white matter injury caused by blood products is raised both by experimental studies and by the demonstration that the presence of IVH is associated with a sharply increased risk of ultrasonographic correlates (e.g., echolucencies) of white matter injury. In one such study, cystic periventricular leukomalacia by ultrasonography was accompanied by IVH in 67% of cases versus only 17% in infants without the cystic injury. A later correlate of white matter injury, nonprogressive ventriculomegaly, is also associated especially with impaired cognitive function in infants with IVH (see Chapter 16 ). Moreover, the neurodevelopmental outcome of preterm infants with later ventricular dilation was worse in those who had associated IVH versus those who did not. Finally, infants with only mild degrees of IVH exhibit a poorer neurodevelopmental outcome than infants with no IVH. The most likely mechanism of white matter injury with intraventricular or (parenchymal blood) involves increased free radical formation , provoked perhaps in part by ischemia-reperfusion but also particularly by local release of iron from the blood. Supportive of this suggestion is the demonstration that non-protein-bound iron was found in the CSF of 75% of preterm infants with posthemorrhagic ventriculomegaly for many weeks after the IVH. Of particular importance in this context are the recent observations of the crucial role of free radicals in the pathogenesis of cerebral white matter injury in the premature infant (see Chapter 15 ).


Selective Neuronal Necrosis


Selective neuronal necrosis , secondary to hypoxia-ischemia in the premature infant, particularly involves the pons, deep nuclear structures, especially the thalamus and basal ganglia, and hippocampus (see Chapter 14 ). Although each of these lesions is more commonly encountered in association with IVH, the relationship is particularly notable for pontine neuronal necrosis. In two carefully studied neuropathological series, 46% and 71% of infants with IVH exhibited pontine neuronal necrosis. Accompanying neuronal necrosis in the subiculum of the hippocampus is common but not invariable. a


a References .

All the infants with IVH accompanied by pontine neuronal necrosis in the series of Armstrong and co-workers died of respiratory failure; previous investigations had suggested that the pontine lesion is related to hypoxic-ischemic insult, hyperoxia, and hypocarbia. Involvement of the inferior olivary nucleus often accompanies the pontine disturbance, and thus cerebellar afferent systems are often affected. This involvement could contribute to the decreased volume of cerebellum observed by volumetric MRI in infants after severe IVH.




Pathogenesis


The pathogenesis of IVH is considered best in terms of intravascular, vascular, and extravascular factors. Clearly, the pathogenesis of IVH is multifactorial, and to some extent different combinations of these factors are operative in different patients. Nevertheless, several of the factors are important in every patient, as discussed in the following sections.


Intravascular Factors


Intravascular factors are those that relate primarily to the regulation of blood flow, pressure, and volume in the microvascular bed of the germinal matrix ( Table 24.4 ). Factors that relate to platelet-capillary function and to blood clotting capability may play a contributory pathogenetic role in certain patients.



TABLE 24.4

Pathogenesis of Germinal Matrix–Intraventricular Hemorrhage: Intravascular Factors

































Fluctuating cerebral blood flow
Ventilated preterm infant with respiratory distress syndrome
Increase in cerebral blood flow
Systemic hypertension: importance of pressure-passive circulation
Rapid volume expansion
Hypercarbia
Decreased hematocrit
Decreased blood glucose
Increase in cerebral venous pressure
Venous anatomy: U-turn in direction of venous flow
Labor and vaginal delivery
Respiratory disturbances
Decrease in cerebral blood flow (followed by reperfusion)
Systemic hypotension: importance of pressure-passive circulation
Platelet and coagulation disturbance


Fluctuating Cerebral Blood Flow


The major importance of fluctuating cerebral blood flow in the pathogenesis of IVH was shown in a study by Perlman and co-workers of ventilated preterm infants with respiratory distress syndrome. Using the Doppler technique at the anterior fontanelle to insonate the pericallosal branch of the anterior cerebral artery (the latter an important source of blood supply to the germinal matrix), we asked whether alterations in cerebral blood flow velocity in the first hours and days of life could be identified and related to the subsequent development of IVH. The findings were decisive. Two patterns of cerebral blood flow velocity were noted on the first day of life: a stable pattern and a fluctuating pattern ( Fig. 24.17 ). The stable pattern was characterized by equal peaks and troughs of systolic and diastolic flow velocity (see Fig. 24.17A ). In contrast, the fluctuating pattern was characterized by marked, continuous alterations in both systolic and diastolic flow velocities (see Fig. 24.17B ); blood flow velocity tracings closely reflected similar patterns of arterial blood pressure, simultaneously obtained from the abdominal aorta through an umbilical artery catheter (see Fig. 24.17 ). A striking relationship of the fluctuating pattern of cerebral blood flow velocity to the subsequent occurrence of IVH was defined when the infants were studied by serial cranial ultrasound scans ( Table 24.5 ).




Figure 24.17


Cerebral blood flow velocity in ventilated premature infant with respiratory distress syndrome.

The upper trace of each pair is the cerebral blood flow velocity, obtained at the anterior fontanelle, and the lower trace is the simultaneous blood pressure obtained with an umbilical artery catheter. (A) Stable pattern and (B) fluctuating pattern. See text for description.


TABLE 24.5

Relation of Fluctuating Cerebral Blood Flow Velocity to Subsequent Development of Intraventricular Hemorrhage
















CEREBRAL BLOOD FLOW VELOCITY PATTERN SUBSEQUENT IVH NO IVH
Fluctuating 21 2
Stable 7 a 20

IVH , Intraventricular hemorrhage.

a Other provocative factors (e.g., pneumothorax) present in four patients.



The aforementioned observations were important for two reasons: First, they identified a subset of infants with respiratory distress syndrome at extreme risk for the subsequent occurrence of IVH and therefore prime candidates for preventive interventions (see later discussion). Second, they suggested a rational pathogenetic mechanism for the development of IVH with the respiratory distress syndrome (i.e., continuous fluctuations of blood flow in the vulnerable matrix microvessels, leading to rupture of these vessels). The relationship of the fluctuating phenomena to the hypoperfusion-reperfusion cycles discussed later is striking. The relationship between fluctuating cerebral blood flow velocity and occurrence of major IVH was later confirmed. Two studies in which fluctuations in flow velocity were less than 10% (coefficient of variation) did not show a correlation of fluctuations with the occurrence of IVH, consistent with the earlier observation of Perlman and co-workers that fluctuations of this small degree do not lead to IVH.


The cause of the fluctuations in both the systemic and cerebral circulations is related to the mechanics of ventilation and to primary and secondary cardiovascular effects, including lowered cardiac stroke volume and cardiac output, often combining effects on both systemic and cerebral blood flows from cardiovascular and ventilatory management ( Fig. 24.18 ). Thus hypercarbia, hypovolemia, hypotension, restlessness , patent ductus arteriosus, and relatively high inspired oxygen concentrations have all correlated with the occurrence of fluctuations in cerebral blood flow velocity.




Figure 24.18


Patterns of regional cerebral saturation (rScO 2 ) and near arterial blood pressure during neonatal illness and procedures.

(A) Patterns of rScO 2 and near arterial blood pressure during and after closure of a hemodynamically important patent ductus arteriosus (PDA) in a 28-week-old preterm neonate. Note the sometimes unusually low rScO 2 during PDA (1), the increase of rScO 2 after the start of indomethacin treatment (2), and the “normalization” of rScO 2 values after ductal closure (3). (B) Pattern of rScO 2 during high-frequency oscillation with high mean airway pressures (MAP: 18 cm H 2 O) and subsequent lowering (red arrow) to 14 cm H 2 O. Note the rather low rScO 2 values (53%) and its increase to reference values after adjustment (lowering) of the MAP. (C) Simultaneous patterns of SaO 2 and rScO 2 before, during, and after recurrent apneas in an unstable preterm neonate. Note the quick recovery of rScO 2 after the first apnea is treated with extra oxygen (1, 2) and the subsequent apnea is treated with extra oxygen (3, 4), showing extremely high rScO 2 levels, up to 94%. (D) Patterns of rScO 2 and mean arterial blood pressure (MABP). Note the similar patterns of rScO 2 and MABP in the upper part (blood pressure passive cerebral oxygenation) and the stable rScO 2 , despite large swings in MABP from 24 to 40 mm Hg, in the lower part (blood pressure–independent cerebral circulation).

(From van Bel F, Lemmers P, Naulaers G. Monitoring neonatal regional cerebral oxygen saturation in clinical practice: value and pitfalls. Neonatology. 2008;94:237–244.)


Increases in Cerebral Blood Flow: Importance of Pressure-Passive Circulation


The close temporal correlation between the occurrence of IVH and abrupt increases in arterial blood pressure, apparent cerebral blood flow (jugular venous occlusion plethysmography), and cerebral blood flow velocity has supported the earlier suggestion that increases in cerebral blood flow play an important pathogenetic role in IVH. A particularly likely cause of the premature infant’s apparent propensity for dangerous elevations of cerebral blood flow is a pressure-passive state of the cerebral circulation . As discussed in Chapters 13 and 15 , severely impaired cerebrovascular autoregulation was identified in approximately 50% of ventilated very low birthweight infants studied by near-infrared spectroscopy in the first several days of life. Using a more sophisticated approach with the same methodology, Soul and co-workers showed that fully 87 of 90 infants studied in the first 5 days of life had pressure-passive periods, and for the total group, these periods accounted for a mean of 20% of the time. Indeed, some infants exhibited the pressure-passive state more than 50% of the time. In addition, hypercarbia and perhaps decreased hematocrit or decreased blood glucose may contribute to severe enough elevations in cerebral blood flow in the premature infant to provoke IVH (see later discussion).


A more complex interaction of the cardiovascular system—both systemic and cerebral—has recently been explored by simultaneous study of neonatal echocardiography and near infrared spectroscopy. In a prospective study of 22 preterm infants between 23 and 27 weeks studied in the first 3 days of life, different patterns of changes in hemodynamics were found in very preterm neonates who developed high-grade IVH compared with those who did not. Importantly, in the infants of the grade IV IVH group, the changes in systemic and cerebral hemodynamics preceded the occurrence of the bleeding and revealed a pattern consistent with a hypoperfusion-reperfusion cycle. More specifically, the infants in the grade IV IVH group had lower cardiac stroke volume and mean blood pressure on study entry at around 6 hours of life. In addition, they also had lower cerebral rSO 2 and higher cerebral functional oxygen extraction during the first 12 hours of monitoring, suggestive of initial low cerebral blood flow in patients. Importantly, this period of hypoperfusion was followed by increased cardiac stroke volume and evidence of cerebral reperfusion , all of which preceded the radiological recognition of IVH. The high concordance in timing and measures of systemic and cerebral vascular changes are consistent with systemic and cerebral hypoperfusion-reperfusion cycles, apparently important in the causative pathway to high-grade IVH.


The importance of cerebral hypoperfusion-reperfusion cycles is also emphasized in a study using superior vena cava (SVC) flow as a surrogate for systemic blood flow and showing that most cases of high-grade IVH were first noted after low SVC flow normalized. A recent case-control study using NIRS also found higher cerebral rSO 2 and lower cerebral oxygen extraction values before the occurrence of high-grade IVH. However, this study did not document the initial cerebral ischemic period and did not monitor systemic hemodynamics. As the systemic and cerebral ischemic period is transient, and because in the study just cited the cranial ultrasound studies were performed on average only every 21 hours, it is conceivable that the period of cerebral ischemia was missed. The underlying primary cause or causes of the cerebral ischemia leading to predisposition to the development of high-grade IVH is/are not known. Myocardial immaturity, with an increased sensitivity to afterload, has been postulated as one of the primary etiological factors of the decreased cardiac output and resultant low cerebral blood flow in the very early hours following delivery of the very preterm newborn. Such findings require further confirmation to propose rational consideration of interventions to prevent a primary and important period of cerebral hypoperfusion. The potential sequence of cardiovascular changes is outlined in Fig. 24.19 .




Figure 24.19


Mechanisms of cerebral ischemia and reperfusion in the pathogenesis of germinal matrix–intraventricular hemorrhage (IVH). PDA, Patent ductus arteriosus.


Elevations of Arterial Blood Pressure and Pressure-Passive Cerebral Circulation.


Concerning the role of elevations in arterial blood pressure, the presence of a pressure-passive cerebral circulation would be expected to lead to an increase in cerebral blood flow in association with increases in blood pressure, with the potential consequence being rupture of vulnerable germinal matrix vessels. The striking increase in cerebral blood flow associated with increases in blood pressure can be shown in real time by near-infrared spectroscopy ( Fig. 24.20 ). A decisive demonstration of the relation between pressure-passive cerebral circulation and the occurrence of IVH was obtained from a classic study of 57 preterm infants supported by mechanical ventilation during at least the first 48 hours of life ( Fig. 24.21 ). Infants in whom ultrasonographic signs of severe IVH developed had prior evidence of a pressure-passive cerebral circulation, whereas those with intact cerebrovascular autoregulation developed either no hemorrhage or only mild hemorrhage (see Fig. 24.21 ). The work of Tsuji and co-workers showed that 47% of infants with impaired cerebrovascular autoregulation developed IVH (or periventricular leukomalacia, or both), whereas only 13% of those with intact autoregulation developed these lesions. Consistent with a potential role for arterial hypertension in this setting is the demonstration of a relationship between maximum systolic blood pressure above a threshold value and subsequent occurrence of IVH. The limit for the highest tolerable peak systolic blood pressure was markedly lower for the smaller infants. A particular role for minute-to-minute alterations in blood pressure has also been demonstrated.




Figure 24.20


Changes in blood pressure (mean arterial pressure [MAP]) and cerebral perfusion (hemoglobin difference [HbDiff]) during a diaper change.

Simultaneous tracings were obtained from a premature infant (30 weeks of gestational age). Note the marked, parallel increase in cerebral perfusion, determined by near-infrared spectroscopy (NIRS), and in arterial blood pressure, obtained from an umbilical artery catheter.

(Courtesy Dr. Adre du Plessis.)



Figure 24.21


Cerebral blood flow (CBF) —mean arterial blood pressure (MABP) reactivities (percentage of change in CBF per millimeter of mercury change in MABP) in premature infants before intracranial hemorrhage.

CBF-MABP reactivities were obtained in the first 2 days of life (primarily in the first 24 hours) in 57 mechanically ventilated preterm infants who had normal ultrasound scans at the time of the reactivity measurements and who were followed subsequently by ultrasonography. Groups A, B, and C were determined by the results of the subsequent scans. The average reactivity and 95% confidence limits for each group are shown. Intact autoregulation (i.e., zero value for CBF-MABP reactivity) was present in those infants who had subsequent scans that were normal or showed only mild hemorrhage. Infants who later developed severe hemorrhage had a pressure-passive cerebral circulation.

(Redrawn from Pryds O, Greisen G, Lou H, Friis-Hansen B. Heterogeneity of cerebral vasoreactivity in preterm infants supported by mechanical ventilation. J Pediatr . 1989;115:638–645.)


A more recent study continued to suggest the importance of elevations in cerebral perfusion being associated with high-grade IVH. Thus cranial Doppler studies for middle cerebral artery cerebral blood flow velocity in 185 preterm infants who were receiving mechanical ventilation showed that severe IVH (grades 3 to 4) was associated with an elevation in diastolic closing margin—a measure of cerebral perfusion in diastole that exceeds “cerebral closing margin.” The measures were a complex combination of assumptions based on Doppler-based estimations of cerebrovascular resistance and compliance. This modeling requires replication, but the findings suggest that high-grade IVH was associated with excessive cerebral perfusion. The timing of this elevation in relation to the timing of the IVH was not delineated within the study.


Moreover, as discussed in Chapter 13 , the upper limit of the normal autoregulatory range in the infant is dangerously close to the upper limit of the range of normal blood pressure. Studies in developing animals indicate that the receptor number for specific vasoconstricting prostaglandins, which are important in setting the upper limit of the autoregulatory range in the adult, are low early in maturation and thereby impair protection of the cerebral circulation from increases in blood pressure.


Whether the pressure-passive cerebral circulatory state relates to dysfunctional autoregulation per se, to maximal vasodilation caused by hypercarbia or hypoxemia (or both), to the cranial trauma of even a normal vaginal delivery, to dopamine therapy for hypotension, or to normal arterial blood pressures in the premature infant that are dangerously close to the upslope of a normal autoregulatory curve remains unclear. Experimental support for these several possibilities is available (see Chapter 13 ). Whatever the mechanism, however, the balance of current data imparts particular importance to events that cause elevations in arterial blood pressure, especially abrupt elevations, in the small premature infant.


Causes of Increased Arterial Blood Pressure in the Human Newborn.


The causes of abrupt elevations in arterial blood pressure sometimes shown to be accompanied by increased cerebral blood flow velocity by the Doppler technique, or increased cerebral blood volume by near-infrared spectroscopy in the premature infant, are clearly important to detect (and to prevent, whenever possible; Table 24.6 ). These causes include the following: such physiological events as rapid eye movement (REM) sleep and the first minutes and hours after birth; such caretaking concomitants as inadvertent noxious stimulation, abdominal examination, handling (see Figs. 24.10 and 24.22 ), instillation of mydriatics, and tracheal suctioning ( Fig. 24.23 ); systemic complications such as pneumothorax and rapid infusion of colloid; and neurological complications such as seizures.



TABLE 24.6

Major Causes of Increased Blood Pressure or Cerebral Blood Flow in the Premature Infant a































Related to “physiological” events
Postpartum status
Rapid eye movement sleep
Related to caretaking procedures
Noxious stimulation
Motor activity: spontaneous or with handling
Tracheal suctioning
Instillation of mydriatics
Related to systemic complications
Pneumothorax
Rapid volume expansion: exchange transfusion, other rapid colloid infusion
Ligation of patent ductus arteriosus
Related to neurological complications
Seizure

a See text for references.




Figure 24.22


Increases of arterial blood pressure in the small premature infant.

Continuous recording of mean aortic pressure in a 20-hour-old premature infant weighing 880 g. Note the marked and sustained increase with manipulation. The infant subsequently developed an intraventricular hemorrhage.

(From Lou HC, Lassen NA, Friis-Hansen B. Is arterial hypertension crucial for the development of cerebral haemorrhage in premature infants? Lancet. 1979;1:1215–1217.)



Figure 24.23


Changes in blood pressure with tracheal suctioning in premature infants.

Note the increase in blood pressure that accompanied suctioning in all but one infant.


Although the degree to which these events contribute to the pathogenesis of IVH requires further quantitation and probably depends on concomitant clinical circumstances, particular importance can be attributed to pneumothorax . In one earlier study of nine infants, pneumothorax was accompanied consistently by abrupt elevations of systemic blood pressure and cerebral blood flow velocity, and these circulatory changes were followed within hours by IVH. Studies in newborn dogs documented abrupt increases in arterial blood pressure on rapid evacuation of pneumothorax. Thus both clinical and experimental data emphasize the potentially deleterious circulatory effects of neonatal pneumothorax.


The complexity of interaction between respiratory and cardiovascular factors in the pathway to IVH is also notable with regard to pneumothorax. A major reduction in the risk of pneumothorax occurred following the administration of exogenous surfactant therapy. Indeed, the administration of surfactant reduced the risk of pneumothoraxes by almost 50% (RR 0.63; 95% CI, 0.53 to 0.75). However, despite this reduction in pneumothorax with surfactant administration, there has been no reduction in the incidence of IVH. One possible explanation for this lack of reduction in IVH may relate to changes in cardiovascular and respiratory stability during surfactant administration. As early as 1992, it was noted that during surfactant administration adverse changes in systemic and cerebral oxygenation could be seen. This has been replicated with less severe impact in recent years, with 62% of premature infants displaying reductions in cerebral electrophysiological activity with intubation and surfactant administration. Thus surfactant administration may have a double-edge effect with a positive impact, with reduction of pneumothorax being offset by a potential negative effect of reduced cerebral perfusion during its administration. To determine the impact of therapies during this period of physiological instability in the preterm infant, monitoring of cerebral perfusion can provide much-needed guidance.


Relevant Experimental Studies: Role of Hypertension.


The particular importance of abrupt increases in systemic blood pressure, and cerebral blood flow in pathogenesis has been demonstrated conclusively in elegant experimental studies in the newborn beagle puppy and in the preterm sheep fetus. The newborn puppy, which has been studied most extensively, has a subependymal germinal matrix approximately comparable to that of the human premature infant of 30 to 32 weeks of gestation. Germinal matrix hemorrhage–IVH is produced most readily in this animal by a sequence of hypotension and hypertension produced by blood removal and volume reinfusion ( Fig. 24.24 ). The marked increase in germinal matrix flow provoked by hypertension has been demonstrated strikingly by autoradiography ( Fig. 24.25 ).




Figure 24.24


Intraventricular hemorrhage in the newborn beagle puppy.

Gross intraventricular hemorrhage with dilation of the lateral ventricle (arrow) in cerebrum of a 24-hour-old puppy subjected to hypertension.

(From Goddard J, Lewis RM, Armstrong DL, Zeller RS. Moderate, rapidly induced hypertension as a cause of intraventricular hemorrhage in the newborn beagle model. J Pediatr . 1980;96:1057–1060.)



Figure 24.25


Increase in blood flow to germinal matrix with increase in arterial blood pressure in the newborn dog.

Blood pressure was elevated by infusion of phenylephrine. Blood flow to the germinal matrix was measured by 14 C-iodoantipyrine autoradiography.

(From Pasternak JF, Groothuis DR. Autoregulation of cerebral blood flow in the newborn beagle puppy. Biol Neonate . 1985;48:100–109.)


Rapid Volume Expansion.


The role of rapid volume expansion (see Table 24.4 ) involves not only the administration of blood or other colloid, as described in relation to systemic hypertension, but also the administration of hyperosmolar materials, such as hypertonic sodium bicarbonate. Pressure-passive cerebral circulation may not be the sole or even the principal means by which such infusions may lead to IVH, particularly in the case of sodium bicarbonate. Although the dangers of rapid infusion of hyperosmolar solutions had been noted for many years, an association of IVH in the premature infant administered sodium bicarbonate was emphasized initially by Simmons and co-workers from study of an autopsy population. The association was later confirmed in a CT study of premature infants, and the importance of rapidity of infusion was made apparent. Conflicting reports on the pathogenetic role of sodium bicarbonate relate in part to the failure to take into account such factors as rapidity of administration and also to the problems of extrapolating data to living infants from studies of dead infants, particularly in the case of IVH. At any rate, the mechanism for the effect of rapid infusion of hyperosmolar sodium bicarbonate on intracranial hemorrhage may relate in part to the abrupt elevation of arterial pressure of carbon dioxide (Pa co 2 ) that results in the poorly ventilated or nonventilated patient from the buffering effect of the bicarbonate. The elevated Pa co 2 would then act on cerebral arterioles, by causing an increase in perivascular hydrogen ion (H + ) concentration, to increase cerebral perfusion as outlined next.


Hypercarbia.


The role of hypercarbia in causing increases in cerebral blood flow of pathogenetic importance for IVH may be appreciable in selected infants. Hypercarbia, a common accompaniment of respiratory distress syndrome, respiratory complications, apneic episodes, and so forth, has been demonstrated conclusively to be a potent means for increasing cerebral blood flow in experimental studies (see Chapter 13 ). Indeed, careful studies of mechanically ventilated preterm infants show a pronounced reactivity of cerebral blood flow to changes in Pa co 2 (≈30% increase in cerebral blood flow per kilopascal [kPa] increase in Pa co 2 ) after the first 24 hours of life. Notably, in the first 24 hours of life, this normal reactivity was attenuated markedly (≈10% increase in cerebral blood flow per kPa increase in Pa co 2 ) in mechanically ventilated infants with normal subsequent ultrasonograms, but it was actually absent in infants with subsequent severe IVH. This observation suggested that, in the first day of life at least, hypercarbia of at least a moderate degree may not be a major pathogenetic factor for severe IVH in mechanically ventilated infants. A similar lack of correlation between hypercarbia and IVH is apparent in several other studies. An increased risk for IVH after hypercarbia, however, is suggested in several other reports, including three that used multivariate analysis. In a particularly large study ( n = 463), hypercarbia (defined as Pa co 2 >60 mm Hg) showed a positive relation with IVH. In a later study of permissive hypercapnia to 45 to 55 mm Hg (vs. 35 to 45 mm Hg in the control group) in ventilated premature infants, no statistically significant difference in IVH was noted between the groups, although the incidence of severe IVH was 29% in the permissive hypercapnia group versus 20% in the control group (not statistically significant). Thus a role for hypercarbia in the pathogenesis of IVH may require particularly marked elevations of Pa co 2 . Consistent with this speculation is the demonstration that hypercapnia leads to clearly impaired autoregulation at Pa co 2 levels above 45 mm Hg ( Fig. 24.26 ). Such levels were shown to be significantly associated with the occurrence of severe IVH and periventricular hemorrhagic infarction in a study of 58 infants.




Figure 24.26


Impaired autoregulation with hypercapnia.

Estimated mean slopes and 95% confidence intervals of the autoregulatory plateau for arterial carbon dioxide tension (Pa co 2 ) values 30, 35, 40, 45, 50, 55, and 60 mm Hg for 43 very low birthweight infants. Bars indicate 95% confidence intervals for the mean slopes of the autoregulatory plateaus for Pa co 2 30 ( n = 82), 35 ( n = 94), 40 ( n = 100), 45 ( n = 103), 50 ( n = 100), 55 ( n = 90), and 60 mm Hg ( n = 83). The horizontal line at slope 0 indicates intact autoregulation. The estimated means of the slope of the autoregulatory plateau (cm/sec* mm Hg −1 ) increased as Pa co 2 increased ( P = .004).

(From Kaiser JR, Gauss CH, Williams DK. The effects of hypercapnia on cerebral autoregulation in ventilated very low birthweight infants. Pediatr Res . 2005;58:931–935.)


Decreased Hemoglobin.


The role of decreased hematocrit in causing increases in cerebral blood flow of pathogenetic importance for IVH may be greater than was previously suspected. Thus, as described in Chapter 13 , an inverse correlation exists in the human infant between hemoglobin concentration and cerebral blood flow, as well as between the concentration of adult versus fetal hemoglobin (higher hemoglobin oxygen affinity) and cerebral blood flow. In one study of premature infants in the first days of life, cerebral blood flow increased by 12% per 1-mM decrease in hemoglobin. The inverse relationship between hematocrit and cerebral blood flow described previously in experimental studies has been suggested to result from changes in arterial oxygen content or blood viscosity. Because alterations in newborn hematocrit to less than 60% have little influence on blood viscosity, the major factor in the studies of human infants is considered to be related to arterial oxygen content and thereby cerebral oxygen delivery. Cerebral blood flow presumably increases to maintain cerebral oxygen delivery at a constant level. Consistent with this possibility, apparently stable premature infants with low hematocrits (<21%) had clinically unsuspected high cardiac output. The adaptive response of increased cerebral blood flow may become maladaptive if certain vulnerable capillary beds (e.g., in the germinal matrix) are exposed to the elevated cerebral blood flow. When one considers that iatrogenic blood loss, owing to repeated blood sampling, and low initial blood volume are common in sick premature infants, especially during the periods of highest risk for occurrence of IVH, the role of decreased hematocrit as a cause of IVH could be considerable.


Potentially consistent with supporting the role of anemia in the risk for IVH, a recent study demonstrated an increased risk of severe IVH among VLBW infants following a red cell transfusion within the first 72 hours of birth. This finding remained significant after controlling for confounding variables (RR, 2.02 [95% CI, 1.54 to 3.33]). The authors were unable to determine the underlying mechanism, given the retrospective nature of the review, but one possibility for the increased risk is concomitant anemia requiring transfusion, rather than the transfusion itself. However, the infants developing IVH may have been sicker and thus at greater risk. Indeed, although there was no difference in coagulation measures, the infants with IVH received more frozen plasma and platelet transfusions and had longer ampicillin courses, higher nucleated RBC counts, more vasopressor use, and a higher mortality rate.


Relevant in this context are results of studies of the timing of cord clamping. Delayed cord clamping (DCC; 30 seconds), compared with immediate cord clamping (6 to 7 seconds), is associated with a slightly higher baseline hematocrit (49% vs. 46%), higher mean blood pressure (33.8 vs. 31.9 mm Hg), increased superior vena caval flow, and a significantly reduced risk for IVH (14% vs. 36%). This reduction in IVH with DCC is supported by a meta-analysis of the randomized trials in preterm infants, although it is noteworthy that the largest studies included involve larger preterm infants with birthweights greater than 1500 g. Because of these benefits in preterm infants and the advantages of increasing iron stores in healthy term-born infants, the American College of Obstetricians and Gynecologists recommended a delay in umbilical cord clamping in vigorous term and preterm infants for at least 30 to 60 seconds after birth.


Blood Glucose.


Decreased blood glucose now should be considered in the evaluation of pathogenetic factors for IVH in view of the observation that cerebral blood flow increases twofold to threefold when blood glucose declines to levels lower than 1.7 mM in the premature infant. Blood glucose levels lower than 1.7 mM in premature infants are not unusual in the first days of life in many NICUs (see Chapter 25 ).


Increased blood glucose has also been evaluated as a potential risk factor for IVH. In a recent case-control study of high-grade IVH ( n = 70) compared with no IVH ( n = 108), infants with IVH had significantly more hyperglycemic events (2.9 ± 1.7 vs. 2.4 ± 1.8 events, P < .05) with longer duration (22.2 ± 14.2 vs. 14.1 ± 12.5 hours, P < .001) and a higher hyperglycemic index (1.0 ± 0.9 vs. 1.4 ± 1.0, P = .003). Respiratory distress syndrome, hypotension, and thrombocytopenia increased the adjusted OR for IVH. Hypoglycemia was not independently associated with IVH. Conversely, the increase in hyperglycemic duration most prominently increased the aOR for severe IVH (OR = 10.33; 95% CI, 10.0 to 10.6; P = .033). To avoid hyperglycemia, insulin therapy is often initiated. However, an important randomized controlled trial of tight glycemic control with insulin versus standard care documented a nonsignificant trend toward an increase in the incidence of grade III/IV IVH in the insulin-treated group (insulin 6/38 infants, 14%, vs. standard care 3/43, 7%, P = .35). The insulin-treated group did have more episodes of hypoglycemia. Thus avoidance of protracted hyperglycemia and hypoglycemia may be most prudent as further data are collected on this clinical factor.


Finally, it has been suggested that alterations in the osmotic gradient may occur with hyperglycemia and other metabolic derangements, such as hypernatremia, leading to an increase in the intravascular pressure relative to the surrounding extravascular tissue that may predispose to IVH. Several cohort studies have shown that those states associated with an alteration in the osmotic balance, such as hyperglycemia and hypernatremia (even high sodium intake in the absence of hypernatremia), are associated with an increased risk for IVH. However, the retrospective nature of these studies cannot delineate the underlying mechanism for this increased risk.


Increases in Cerebral Venous Pressure


Elevations of cerebral venous pressure may contribute to the occurrence of IVH. Indeed, the potential importance of venous factors is suggested by the demonstration that with postmortem injection of carotid artery or jugular vein in infants with germinal matrix hemorrhage, the injected material entered the hemorrhage only through venous injections. Moreover, careful anatomical studies also are consistent with an origin at the level of the capillary-venule junction or the small venule. The most important causes for such increases are labor and delivery, asphyxia, and respiratory complications (see later discussion).


Importance of Venous Anatomy.


The particular importance of increased venous pressure in the pathogenesis of IVH relates in part to the venous anatomy in the region of the germinal matrix (see Fig. 24.6 ). Thus the direction of deep venous flow takes a peculiar U-turn in the subependymal region at the level of the foramen of Monro (i.e., the most common site of germinal matrix hemorrhage). Also at this site is the point of confluence of the medullary, thalamostriate, and choroidal veins to form, in sequence, the terminal vein and then the internal cerebral vein, which ultimately empties into the vein of Galen.


Labor and Delivery.


Concerning labor and delivery, marked increases in cerebral venous pressure must be common accompaniments. Indeed, in one study of 46 infants, when measurement of “fetal head compression pressure” was determined by a compression transducer positioned between the fetal head and the wall of the uterus, the overall mean pressure was 158 mm Hg. Deformations of the particularly compliant premature skull are likely to accentuate the increases in venous pressure caused by normal labor. Indeed, the deleterious effects of labor (see later discussion) appear to be most pronounced in the most premature infants. The skull deformations can lead to obstruction of major venous sinuses and presumably increased venous pressure. Support for this notion has been provided by studies of blood flow velocity in the sagittal sinus, cerebral blood volume, and intracranial pressure during such manipulations as external pressure on the skull or rotation of the neck. These effects may be expected to be greater with breech delivery. Available data are somewhat inconsistent concerning a relationship between such factors as presence or absence of labor, duration of labor, mode of delivery, and the occurrence of IVH, although in general the studies were not designed to address these issues specifically and were retrospective. The inconsistency of the data, however, does not rule out a contributory role of intrapartum events in causation of IVH in certain infants. Thus, in a study that addressed the role of presence or absence of labor, duration of labor, mode of labor, and potential confounders in a multivariate analysis, Leviton and co-workers showed that infants delivered vaginally were more likely to develop IVH than those delivered abdominally, that labor longer than 12 hours increased risk of IVH regardless of the mode of delivery, and that the occurrence of labor before abdominal delivery increased the incidence of IVH by 2 to 4 times, depending on the duration of labor ( Table 24.7 ). In a separate study of 201 VLBW infants, multivariate analysis also indicated an increased risk (2.2-fold) of IVH for infants delivered vaginally, a very low risk (7%) for infants delivered abdominally with no labor, and an increased risk among infants delivered abdominally for labor greater than 10 hours in duration (40%). Subsequent investigations of 229 and 254 infants, respectively, show an increased risk of IVH occurring in the first 3 to 12 hours of life as a function of active labor and vaginal delivery. Finally, a multicenter study of 4795 infants of less than 1500 g birthweight showed an incidence of grade III and IV IVH in 19% of vaginally delivered infants and 11% of those delivered by cesarean section without labor. On balance, these data suggest that labor and delivery influence the risk of IVH in premature infants and have implications concerning a potential role for cesarean section in prevention (see later discussion).



TABLE 24.7

Occurrence of Germinal Matrix Hemorrhage as a Function of Route of Delivery and Duration of Labor


























LABOR ROUTE OF DELIVERY
VAGINAL ABDOMINAL
None 6.1% (8/131)
<6 h 23.2% (19/82) 14.7% (12/129)
6–12 h 22.5% (9/40) 18.5% (5/27)
>12 h 32.1% (9/28) 25.0% (3/12)

Data from Leviton A, Fenton T, Kuban KC, Pagano M. Labor and delivery characteristics and the risk of germinal matrix hemorrhage in low birth weight infants. J Child Neurol. 1991;6:35–40.


The most recent studies of mode of delivery relative to IVH have continued to demonstrate conflicting results, with several finding no association with method of delivery. A recent study of 158 infants born at less than 1500 g found that there was an increased risk of mild IVH among infants with vaginal delivery versus cesarean section before the second stage of labor. The studies that did not report an association between mode of delivery and IVH did not comment on the duration of labor or the stage during which the cesarean section was performed, possibly explaining the discrepancies in the literature. Of note, it has been recently shown that head position, both before delivery and in the neonatal nursery, may also alter cerebral venous drainage. One study with near-infrared spectroscopy showed that cerebral venous drainage may be impaired in prone or side positions.


Hypoxic-Ischemic Injury.


With perinatal asphyxial events, circulatory collapse may lead to hypoxic-ischemic cardiac failure and, as a consequence, increased cerebral venous pressure. The cardiac disturbance is caused by injury of papillary muscle, subendocardial tissue, and myocardium. The importance of increased venous pressure in association with asphyxia in the causation of IVH was shown in experimental studies of preterm fetal sheep. Thus it seems likely that increased venous pressure could contribute to the propensity to IVH observed after serious asphyxia. Consistent with this notion are the strong relationships among such factors as severe umbilical cord acidemia, low Apgar scores, the need for neonatal resuscitation, and the occurrence of severe IVH (see later). Other factors associated with asphyxia, such as ischemic injury to the germinal matrix and hypercarbia, are also likely important.


Respiratory Disturbances.


Concerning respiratory disturbances , available data suggest that such factors as positive-pressure ventilation with relatively high peak inflation pressure, tracheal suctioning, abnormalities of the mechanics of respiration, and pneumothorax may be major causes of increased cerebral venous pressure in the premature infant. Thus, extending earlier observations, Cowan and Thoresen used Doppler measurements of blood flow velocity in the superior sagittal sinus to demonstrate a striking sensitivity of the venous circulation to the level of peak inflation pressure; the smallest infants exhibited the most marked effects.


The possibility of a particular importance for venous abnormalities in causation of IVH was raised by a study of intubated preterm infants with respiratory distress syndrome under conditions in which dangerous alterations in arterial blood pressure occur (i.e., elevations with tracheal suctioning and fluctuations with breathing out of synchrony with the ventilator ). The effects on the venous circulation were dramatic. With elevations in arterial blood pressure produced by tracheal suctioning, pronounced changes in venous pressure also occurred ( Fig. 24.27A ). Moreover, because the magnitude and the direction of the changes in venous pressure often were not similar to those in arterial pressure, striking changes in perfusion pressure resulted (see Fig. 24.27B ). Similarly, because fluctuations in arterial blood pressure were associated with noncoordinated fluctuations in venous pressure ( Fig. 24.28A ), pronounced and continuous alterations in perfusion pressure resulted (see Fig. 24.28B ). Thus, under both circumstances, decreases in perfusion pressure by as much as 10 to 20 mm Hg were followed in seconds by abrupt, similar increases in perfusion pressure. Because these changes occur essentially on a beat-to-beat basis, it is unlikely that autoregulation, even if functional, could protect critical capillary beds by causing the changes in arteriolar diameter necessary to maintain constant cerebral blood flow under such circumstances. Thus the previously established role for disturbed mechanics of respiration with fluctuations in arterial blood pressure in the causation of IVH (see Table 24.5 ) may be mediated as much by alterations on the venous side of the cerebral circulation as by alterations on the arterial side. A similar conclusion can be drawn for the previously established role in the causation of IVH of abruptly increased arterial blood pressure with pneumothorax, because this respiratory complication has been shown to cause abruptly increased venous pressure as well. A study of 58 cases of severe IVH with periventricular hemorrhagic infarction showed a significant relationship of pneumothorax with the occurrence of the lesion.




Figure 24.27


Venous pressure response to suctioning.

(A) Simultaneous venous pressure and arterial blood pressure tracings from a premature infant during tracheal suctioning (arrowhead) . Note the pronounced increases in venous pressure after onset of suctioning. (B) Graphic plot of calculated changes in perfusion pressure during the same suctioning episode.

(Adapted from Perlman JM, Volpe JJ. Are venous circulatory abnormalities important in the pathogenesis of hemorrhagic and/or ischemic cerebral injury? Pediatrics . 1987;80:705–711.)



Figure 24.28


Correlation of fluctuations in arterial and venous pressure.

(A) Simultaneous venous pressure and arterial blood pressure tracings from an infant with fluctuating blood pressure. Note that the marked fluctuations in arterial pressure are associated with marked fluctuations in venous pressure. (B) Graphic plot of calculated changes in perfusion pressure in the same infant.

(Adapted from Perlman JM, Volpe JJ. Are venous circulatory abnormalities important in the pathogenesis of hemorrhagic and/or ischemic cerebral injury? Pediatrics . 1987;80:705–711.)


Decreases in Cerebral Blood Flow


Importance of Pressure-Passive Cerebral Circulation.


Decreases in cerebral blood flow, occurring either prenatally (perhaps primarily intrapartum) or postnatally, may play an important role in pathogenesis of IVH in certain infants. The principal consequence of the decreased cerebral blood flow is injury of germinal matrix vessels, which rupture subsequently on reperfusion. The importance of vascular border zones and end zones in the matrix, as well as the intrinsic vulnerability of the matrix vessels to oxygen deprivation, is emphasized later (see the section on vascular factors ). As indicated earlier, hemorrhagic hypotension preceding volume reexpansion is the optimal means to produce IVH experimentally in the newborn beagle puppy. In the premature infant, decreases in cerebral blood flow are most likely with perinatal hypoxia-ischemia and with various postnatal events that result in systemic hypotension. Because of the pressure-passive cerebral circulation in sick premature infants, this hypotension can lead to a decrease in cerebral blood flow. Recall that a detailed study of 90 premature infants in the first 5 days of life showed that more than 95% had pressure-passive periods, with a mean total time of pressure-passivity of 20%.


Perinatal Hypoxic-Ischemic Events.


Although it is not an obligatory event for development of IVH, the infant with prior perinatal asphyxia clearly has an increased likelihood of developing IVH, and, in our experience, the hemorrhage in such infants tends to be relatively large. Indeed, a study of 58 infants with periventricular hemorrhagic infarction found a strong association of the lesion with fetal distress and the need for emergency cesarean section, low Apgar scores, and the need for respiratory resuscitation. Perinatal hypoxic-ischemic events presumably explain, at least in part, the relation between low Apgar scores, early acidosis, early use of bicarbonate or pressors, hypocarbia, and the subsequent overall occurrence of IVH, particularly lesions that develop in the first 12 hours. a


a References .

The mechanism for provocation of IVH with perinatal asphyxia is complex and includes increases in cerebral blood flow associated with impaired vascular autoregulation, increases in cerebral venous pressure, and decreases in cerebral blood flow associated with hypotension, with resulting injury to matrix capillaries. Release of endogenous vasodilators (e.g., adrenomedullin) in the first hours after birth may also play a role in this situation. Studies of the concentrations of brain-specific creatine kinase isoenzymes in cord blood or blood samples obtained early in the postnatal period or of the hypoxanthine metabolite, uric acid, in blood samples obtained on the first postnatal day, in preterm infants who later developed IVH, support the notion that late intrauterine injury (perhaps asphyxia) may be involved in at least some cases of IVH. In one study, those infants who developed IVH had cord blood levels of brain-specific creatine kinase (CK-BB) that were 6 times greater than those in the infants who did not develop IVH. Moreover, the possibility of a perinatal hypoxic-ischemic insult to brain as a predecessor to IVH is suggested also by the finding of depressed amplitude-integrated electroencephalographic activity before the occurrence of the hemorrhage in 4 of 10 carefully studied preterm infants. In a separate study, early continuous electroencephalographic monitoring detected such abnormalities as excessive discontinuity before the occurrence of IVH. These abnormalities are similar to those produced by hypoxic-ischemic insults (see Chapter 10 ).


More recent studies have documented a high incidence of seizures in the preterm infant in the first 72 hours of life, accompanied by a strong association with the presence of IVH. In one study, 95 VPT infants underwent aEEG monitoring during the first 72 hours of life. The overall incidence of seizures in this sample was 48%. High seizure burden was associated with increased risk of IVH throughout each of the 3 days of monitoring. The seizures observed in this very preterm cohort demonstrated a similar evolution in the timeline to the seizures of a term infant suffering from hypoxic-ischemic encephalopathy (HIE) with the highest median seizure burden during the 0- to 24-hour period. These observations support a potential role of perinatal asphyxia in the pathway to IVH.


Postnatal Ischemic Events.


The importance of postnatal decreases in cerebral blood flow in the pathogenesis of IVH has been shown by studies using a combination of noninvasive methods to evaluate the cerebral circulation. Notably, however, a relationship with decreases in mean arterial blood pressure , when infants who developed IVH were compared with infants who did not develop IVH, was not uniformly shown. Thus the changes in cerebral hemodynamics could occur without pronounced disturbances of mean arterial blood pressure . These findings of a lack of association between mean arterial blood pressure and IVH were recently replicated. Rhee and co-workers documented no relationship between systemic blood pressure and risk for IVH but did document a relationship between measures of cerebral blood flow with IVH. This finding continues to emphasize the inability of systemic measures of blood pressure to define the adequacy of cerebral perfusion. Because impaired autoregulation and fluctuations in blood pressure are both frequent but not necessarily constant in the sick preterm infant, intermittent declines in cerebral blood flow could occur without pronounced changes in mean blood pressure. Thus continuous measurements of the cerebral circulation are critical in determining changes in cerebral blood flow in the sick, ventilated infant .


Nevertheless the importance of postnatal decreases in arterial blood pressure in the pathogenesis of IVH is suggested by several studies in which arterial blood pressure was monitored continuously from birth or the first hours of life. In a study of approximately 25,000 VLBW infants, severe IVH was 3 times more common in those infants requiring cardiopulmonary resuscitation (15.3%) at delivery than in those not requiring resuscitation (4.9%). A relationship between arterial hypotension and subsequent occurrence of IVH has been documented frequently. Although the possibility of ischemic insult to germinal matrix in the postnatal period is obvious in association with such occurrences as severe apnea, myocardial failure, sepsis, and so forth, sharp decreases in blood pressure have also been shown to precede the more widely recognized increases provoked by ordinary caretaking procedures. By continuous monitoring of arterial blood pressure during such procedures as auscultation of the chest, taking of temperature, and suctioning, the increases in blood pressure previously noted were shown to be preceded by a decrease in blood pressure. The decreases were most pronounced in the infants requiring the most intensive ventilatory support. The biphasic response of decrease and then increase in blood pressure is qualitatively similar to the sequence required to produce IVH in the beagle puppy (see earlier). The rebound elevation of cerebral blood flow velocity observed after apnea and bradycardia is relevant in this context.


Of particular importance concerning a role for postnatal ischemia is the demonstration by near-infrared spectroscopy in 24 preterm infants in the first 24 hours of life that cerebral blood flow was significantly lower in infants with subsequent demonstration of IVH (median, 7.0 mL/100 g/min) than in those without IVH (median, 12.2 mL/100 g/min). In addition supportive of a relationship between postnatal ischemic events and the occurrence of IVH is the demonstration of decreased cardiac output in the first 36 hours of life in infants who developed the lesion, especially with severe IVH. Similarly, the occurrence of periventricular hemorrhagic infarction with severe IVH was strongly associated with metabolic acidosis in the first days of life and a need for pressor support and volume expanders. Finally, the beneficial effect of delayed versus immediate clamping of the umbilical cord on the incidence of IVH is considered to be likely caused in considerable part by stabilization of cerebral blood flow and prevention of ischemia. However, as mentioned earlier, the interrelationships between cerebral hypoperfusion and IVH appear complex, with an early ischemic period potentially increasing the vulnerability for reperfusion injury to the germinal matrix. These evolutions can only be evaluated with the presence of monitoring of both systemic and cerebral hemodynamics simultaneously.


The contributory role of maternal intrauterine infection and fetal systemic inflammation in the pathogenesis of IVH likely is mediated by effects on the cerebral circulation. Although data are not entirely consistent concerning a relationship between chorioamnionitis and the occurrence of IVH, several reports indicate an association with IVH of elevated levels of specific cytokines, especially interleukin-6 (IL-6), in cord or early neonatal blood. Infants with chorioamnionitis and fetal cord vasculitis had higher IL-6 levels and likelihood of IVH than those with fetal vasculitis alone. The findings suggest that maternal intrauterine infection that leads to a systemic fetal inflammatory response is critical. Indeed, a recent large study of periventricular hemorrhagic infarction ( n = 58) found no association with maternal fever, maternal infection, or pathologically confirmed chorioamnionitis. The principal mechanism of the fetal/neonatal cytokine effect is likely circulatory disturbance. Thus decreased arterial blood pressure, often requiring pressor support, has been shown in the infants with elevated IL-6. IL-6, like several other cytokines, has vasodilator properties that likely lead to the decreased blood pressure and presumably cerebral blood flow ( Fig. 24.29 ). Whether IL-6 or related cytokines impair cerebrovascular autoregulation is plausible but not yet shown.




Figure 24.29


Relationship between mean arterial blood pressure (BP) and interleukin-6 (IL-6) concentration.

Mean BP was inversely correlated ( R 2 = 0.21, P < .01) with log IL-6. Solid square, chorioamnionitis; open square, no chorioamnionitis.

(From Yanowitz TD, Jordan JA, Gilmour CH, Towbin R, et al. Hemodynamic disturbances in premature infants born after chorioamnionitis: association with cord blood cytokine concentrations. Pediatr Res . 2002;51:310–316.)


Platelet and Coagulation Disturbances


Disturbances of platelet-capillary function and coagulation may contribute to the pathogenesis of IVH (see Table 24.4 ). The lack of uniformity in results of studies designed to investigate the pathogenetic role of such disturbances, however, emphasizes that the role is likely to be contributory or important only in certain patients.


Platelet-Capillary Function.


Regarding platelet-capillary function, an earlier prospective study is of particular interest. Forty percent of infants of less than 1500 g birthweight exhibited platelet counts less than 100,000/mm 3 , and most of these thrombocytopenic infants had abnormal bleeding times. The incidences of IVH in thrombocytopenic versus nonthrombocytopenic infants were 78% versus 48% for those weighing less than 1000 g. Additional analysis for other factors potentially important for causation of IVH suggested that the presence of thrombocytopenia was an independent pathogenetic factor. Subsequent work has both confirmed and refuted a role for thrombocytopenia in the pathogenesis of IVH.


While subsequent work has both confirmed and refuted a role for thrombocytopenia in the pathogenesis of IVH, the most recent studies have tended to support an association. The largest of these evaluated 655 infants born at less than 32 weeks’ gestational age, 44% of whom had thrombocytopenia. Within this cohort there was a 30% (85/286) incidence of IVH in those with thrombocytopenia versus 14% (53/369) in those with a normal platelet count. Interestingly there was no correlation between the severity of thrombocytopenia and the incidence of IVH; rather the relationship was dichotomous. Similarly no protective effect was demonstrated with increased platelet transfusion, as might be expected if there was a linear correlation with platelet count. In an alternate cohort of 251 neonates, Rastogi and colleagues found no significant difference in severe IVH with or without thrombocytopenia in isolation, but the odds of a severe IVH were increased fourfold if there was a sudden drop in the platelet count. The odds increased to 14 times the baseline if this sudden decline occurred from a baseline of thrombocytopenia. The exact mechanism between IVH and thrombocytopenia has not yet been elucidated. Some evidence suggests that premature infants demonstrate elevated levels of prostacyclin before the occurrence of IVH. Because prostaglandins have an impact not only on platelet function but also on such factors as cerebral blood flow and free radical production that also may be important in pathogenesis of IVH, it is not clear to what extent effects on platelet-capillary function were independently related to causation of IVH.


Perhaps relevant to the possibility of disturbed platelet function (rather than platelet count) in some infants before the occurrence of IVH is the study by Rennie and co-workers of circulating levels of the principal metabolite of prostacyclin in preterm infants. Prostacyclin is a potent perturbant of platelet-capillary function and is produced in elevated amounts, probably by lung, in respiratory distress syndrome and with mechanical ventilation. Evidence was obtained for elevated levels of prostacyclin before the occurrence of IVH. Because prostaglandins additionally have an impact on such factors as cerebral blood flow and free radical production that also may be important in the pathogenesis of IVH, it is not clear to what extent effects on platelet-capillary function were independently related to causation of IVH. More data are needed on these issues.


Coagulation.


Disagreement exists concerning the role of coagulopathy in causation of IVH. It could be postulated that prothrombotic disturbances (e.g., factor V Leiden mutation, prothrombin mutation) would increase IVH by provoking deep venous thrombosis or would decrease IVH by preventing extension of small hemorrhages. Because disturbances of coagulation are not uncommon in preterm infants with other provocative factors for IVH (e.g., serious respiratory distress syndrome, asphyxia) or may occur secondary to major hemorrhage, an independent pathogenetic role for such disturbances has been difficult to establish. Although administration of fresh frozen plasma was shown in one study to decrease the incidence of IVH, no effect on coagulation measures accompanied the apparent beneficial effect. Moreover, a later investigation failed to show a beneficial effect of prophylactic early fresh frozen plasma in premature infants. Administrative of antithrombin III, known to be low in premature infants, did not alter the incidence of IVH in a randomized study.


Recent reports have examined the potential role of mutations in coagulation proteins as possible risk modifiers for IVH. Several common mutations in coagulation proteins are associated with increased tendency to thrombotic events. Those that have been investigated most included factor XIII Val34Leu mutation, factor V Leiden, factor II (prothrombin) G20210A, and MTHFR C677T. Factor XIII stabilizes the fibrin clot, and thus it is hypothesized that low levels of factor XIII may predispose to IVH. Low factor XIII levels in preterm infants may contribute to the relatively higher fibrinolytic activity documented in preterm infants. Two recent studies, however, failed to find any association between factor XIII-Val34Leu mutation and IVH. Factor V Leiden is a common mutation in the white population and is associated with thrombotic events, with decreased inactivation of factor V leading to increased thrombin generation. The role of factor V Leiden in risk of IVH is unclear. Results have been conflicting with some studies suggesting a significant role for factor V Leiden in the risk of IVH, while others suggest no role, or a mixed pattern, whereby the mutation is associated with low-grade IVH (grade I or II), but a reduced risk of severe IVH.


There have been similarly conflicting findings for prothrombin G20210A mutations and the development of IVH. Two mutations (C677T and A1298C) in the 5,10-methylene tetrahydrofolate reductase (MTHFR) gene lead to hyperhomocysteinemia under conditions of decreased folate or B 12 concentrations and an increased risk of thrombosis. Although no consistent association has been found with this mutation and IVH, a recent report by Ment and co-workers did find a significantly increased association of this polymorphism among preterm infants, specifically with grade II–IV IVH.


Potential Role of Drugs.


The possibility that maternal ingestion of certain drugs , such as aspirin, can result in impaired neonatal hemostasis and provoke IVH was suggested by two earlier reports. It is not likely, however, that such factors play a major independent role. Similarly, retrospective data suggest that the use of heparin as an intravascular flush to maintain patency of umbilical artery catheters increases the risk of IVH by fourfold. Whether this effect is related to a disturbance of coagulation or to selection bias because of the use of heparin in sick premature infants who develop IVH for other reasons is unclear. A subsequent report also suggested an increasing risk for IVH in premature infants as a function of the daily dose of heparin. More data are needed.


Vascular Factors


Vascular factors are those referable to the blood vessels of the germinal matrix ( Table 24.8 ). As discussed earlier (see the section on neuropathology ), the vascular site of origin likely involves the rich microcirculation of the germinal matrix and, specifically, endothelial-lined vessels not readily characterized as arterial or venous. Thus vascular factors are best grouped in two categories—those suggesting (1) that the integrity of small matrix vessels is tenuous and (2) that these vessels are particularly vulnerable to hypoxic-ischemic injury (see Table 24.8 ).



TABLE 24.8

Pathogenesis of Germinal Matrix–Intraventricular Hemorrhage: Vascular Factors

















Tenuous capillary integrity
Involuting, remodeling capillary bed
Deficient vascular lining
Large vascular and luminal area
Vulnerability of matrix capillaries to hypoxic-ischemic injury
Vascular border zones
High requirements for oxidative metabolism


Tenuous Vascular Integrity


Three lines of anatomical evidence suggest that the integrity of the microvasculature is tenuous in the germinal matrix (see Table 24.8 ). First, these vessels, like the germinal matrix itself, are in a process of involution. Pape and Wigglesworth characterized the elaborate capillary bed of the germinal matrix as “a persisting immature vascular rete,” an immature microvascular network that is remodeled into a mature capillary bed when the matrix disappears. In keeping with this notion, transmission electron microscopic studies of the matrix reveal many small vessels with the absence of a complete basal lamina, a fenestrated lining, and other features characteristic of immature vessels. This involuting remodeling capillary bed may be expected to be more susceptible to rupture than would more mature vessels.


Second, many studies emphasized that the matrix microcirculation is composed of simple endothelial-lined vessels, often of a larger size than capillaries but not readily categorized as arterioles or venules because of absence of muscle and collagen. Particularly careful studies have documented in detail the absence of muscle and of type VI collagen. Investigators have postulated that such vessels are likely to be more susceptible to rupture. In favor of this postulate is the demonstration in the newborn beagle puppy of matrix vessels that are similar to those just described (i.e., relatively large size and thin walls ); these vessels are the sites of hemorrhage in this animal model of germinal matrix hemorrhage–IVH. Recent work emphasized the high vascular density of the germinal matrix and the abundance of small veins, the walls of which are lined only by endothelial cells.


Third, an electron microscopic study of cortical and germinal matrix vessels showed a twofold to fourfold greater diameter of both the vessels and the lumina of the vessels of the germinal matrix versus those of the cortical plate in infants of 25 to 33 weeks of gestation. At 37 weeks of gestation, this difference had disappeared and, notably, the diameters of the matrix vessels and their lumina were twofold to threefold smaller than at 25 to 33 weeks. Thus, in the age range of greatest propensity for occurrence of IVH, the diameters are unusually large, a finding of potential pathogenetic importance because of the Laplace law, which states that the larger the vessel diameter, the greater the total force on the wall at any given pressure.


Fourth, as noted earlier, additional potential characteristics leading to fragility of the vasculature include discontinuous glial endfeet, relative lack of pericytes, and immature basal lamina components, among other features. Exuberant angiogenesis, postulated to be related partially to relative hypoxia of the richly cellular matrix and manifested by high VEGF and angiopoietin-2 levels, may lead to fragility and relative ease of rupture (see the earlier section on the site of origin).


The possibility should be considered that the effects of these maturational deficiencies of germinal matrix vessels underlie the interesting observation that women with a diagnosis of preeclampsia exhibit a lower risk for an infant with IVH (2.5%) than women without this diagnosis (17%). This lower risk for IVH has been confirmed. Infants born to preeclamptic mothers exhibit a variety of features suggestive of accelerated maturation of the brain and other organs.


Vulnerability to Hypoxic-Ischemic Injury


Two features suggest a particular vulnerability of matrix capillaries to hypoxic-ischemic injury (see Table 24.8 ). First, as shown by Takashima and Tanaka, at the usual site for matrix hemorrhage a vascular border zone exists between the end fields of the striate and thalamic arteries ( Fig. 24.30 ). The demonstration of heterogeneity of blood flow within the matrix of the beagle puppy may represent the physiological correlate of the anatomical data. Studies of hypotension in the fetal rat also demonstrate a vulnerability of the matrix to ischemia. Thus it can be postulated that the matrix vessels could be readily injured by ischemia, and on reperfusion of these injured vessels, hemorrhage could occur. As indicated earlier, this notion is supported by the finding in the beagle puppy model of IVH that the free radical scavenger superoxide dismutase prevents hemorrhage caused by the usual hypotension-reperfusion sequence. Similarly, certain older studies in human infants (see later discussion) suggest that vitamin E, a free radical scavenger, has a preventive effect on IVH.




Figure 24.30


Vascular border zone and end zone at the site of germinal matrix hemorrhage.

Postmortem arteriography of cerebral hemisphere (coronal section) of a premature infant of 32 weeks of gestational age. Note the triangular avascular area (large arrow) at the border zone between the end fields of the thalamostriate and medullary arteries at the site of a small subependymal hemorrhage (small arrow) .

(From Takashima S, Tanaka K. Microangiography and vascular permeability of the subependymal matrix in the premature infant. Can J Neurol Sci . 1978;5:45–50.)


Second, matrix capillaries, like other brain capillaries, appear to have a high requirement for oxidative metabolism. Thus brain endothelial cells have been shown to contain 3 to 5 times more mitochondria than systemic capillary endothelial cells. This intense oxidative activity would complement the presence of a vascular border zone in the matrix in enhancing the vulnerability of matrix vessels to ischemic insults.


Extravascular Factors


Extravascular factors are those referable to the space surrounding the germinal matrix capillaries ( Table 24.9 ). These factors are grouped best into the following three categories: deficient vascular support, fibrinolytic activity, and postnatal decrease in extravascular tissue pressure.



TABLE 24.9

Pathogenesis of Germinal Matrix–Intraventricular Hemorrhage: Extravascular Factors








  • Deficient vascular support



  • Fibrinolytic activity



  • Postnatal decrease in extravascular tissue pressure (?)



Deficient Vascular Support


The germinal matrix can be seen by gross examination to be a gelatinous, friable structure, and by microscopic examination to be deficient in supporting mesenchymal and glial elements. Thus investigators have suggested that the extravascular space provides poor support for the large, endothelial-lined capillaries that course through it and that are the site of the hemorrhage. This formulation received experimental support from studies of the germinal matrix of the beagle puppy, which showed that large portions of the capillary walls lacked any direct contact with perivascular structures (unlike the capillaries in the cerebral cortex or caudate nucleus). Decisive demonstration of the potential importance of deficient vascular support in the human infant is derived from the work of Gould and Howard. Thus, assessing astrocytic fibrillary development by immunocytochemical staining for glial fibrillary acidic protein (GFAP), these investigators showed minimal astrocytic development as late as 27 weeks of gestation and prominent GFAP staining, not until 31 weeks. A later detailed study also showed deficient GFAP and astrocytic endfeet overlying the germinal matrix vasculature during the gestational period of 23 to 34 weeks. Because of the role of glial fibers in capillary stabilization, these data suggest an important role for deficient astrocytic development in pathogenesis of IVH in the premature infant. Lastly, immunomicroscopy of neuropathology specimens have shown that staining for tight junction proteins (ZO-1, claudin, and occludin) in the germinal matrix vessels at 24 weeks revealed reduced and immature staining patterns. Immature tight junctions between endothelial cells would affect the functionality of the blood-brain barrier and could predispose the vessels to hemorrhagic rupture.


Fibrinolytic Activity


An excessive amount of fibrinolytic activity has been defined in the periventricular, germinal matrix region of the human premature infant (see Table 24.9 ). Although the source of this activity is not established conclusively, it is likely that the fibrinolytic activity reflects the proteolytic action of the plasmin-generating system. This extracellular proteolytic system is composed of plasminogen activator, plasminogen, and plasmin. Plasminogen activator, a protease secreted from cells, activates plasminogen to generate the protease plasmin, which, in turn, can degrade a wide variety of extracellular proteins. This system is involved in many developing, remodeling tissues as a normal maturational process. Fibrinolysis is only one action of this proteolytic system. In developing chick spinal cord, this system is most active during glial proliferation. Moreover, studies in cell culture show that the immature astrocyte (not the mature astrocyte) is the principal source for plasminogen activator and the action of this proteolytic system. Glial proliferation is very active in the germinal matrix at the usual time of occurrence of germinal matrix hemorrhage. It is reasonable to suspect that this fibrinolytic activity, an epiphenomenon of the proteolytic system required for remodeling of the germinal matrix, allows the small capillary hemorrhages of the matrix to become the large lesions characteristic of IVH.


Postnatal Decrease in Extravascular Tissue Pressure


The possibility that a postnatal decrease in extravascular tissue pressure causes an increase in the transmural intravascular-extravascular pressure gradient sufficient to provoke hemorrhage was raised by studies of the beagle puppy. Administration of prolactin was shown to decrease the incidence and severity of hemorrhage in that animal model. The relevance to hemorrhage in the human premature infant, however, is unclear because the timing of the apparent postnatal decrease in extracellular volume in the premature infant (i.e., 2 to 3 days of life) occurs after the peak time of occurrence of IVH. As noted earlier, it has been suggested that alterations in the osmotic gradient may occur with hyperglycemia and other metabolic derangements, such as hypernatremia, leading to an increase in the intravascular pressure relative to the surrounding tissue that may predispose to IVH. Several cohort studies have shown that these states associated with an alteration in the osmotic balance, such as hyperglycemia and hypernatremia (even high sodium intake in the absence of hypernatremia), are associated with an increased risk for IVH. However, the retrospective nature of these studies cannot delineate the underlying mechanism for this increased risk.


Interaction of Pathogenetic Factors


As indicated throughout the preceding discussion, not all the pathogenetic factors operate in every case. Clinical circumstances dictate which factors are most critical in the individual infant . Perhaps the best example of the interaction of the most important pathogenetic factors is provided by the clinical situation of the premature infant who is mechanically ventilated for serious respiratory distress syndrome (see Fig. 24.19 ), the clinical setting for the largest proportion of all cases of IVH.




Clinical Features


The principal clinical setting for IVH is a premature infant with respiratory distress syndrome severe enough to require mechanical ventilation. The time of onset of hemorrhage , defined most clearly by serial cranial ultrasonography (see the section on diagnosis ), is the first day of life in at least 50% of affected infants, and by 72 hours, approximately 90% of the lesions can be identified. The timing of the initial clinical features, as expected, is similar.


Three Basic Syndromes


Three basic clinical syndromes accompany IVH: (1) a catastrophic deterioration ( Table 24.10 ), (2) a saltatory deterioration ( Table 24.11 ), and (3) a clinically silent syndrome. The least common but most dramatic of these is the catastrophic deterioration, which occurs in infants with the most severe hemorrhages. More common is the saltatory deterioration, and most common of all is the clinically silent syndrome . The latter two syndromes occur most often, although not exclusively, in infants with smaller lesions.



TABLE 24.10

Catastrophic Clinical Syndrome With Germinal Matrix–Intraventricular Hemorrhage








  • Inexorable evolution in minutes to hours



  • Neurological features




    • Stupor → coma



    • Respiratory disturbance → apnea



    • Generalized tonic seizures



    • “Decerebrate” posturing



    • Pupils fixed to light



    • Eyes fixed to vestibular stimulation



    • Flaccid quadriparesis




TABLE 24.11

Saltatory Clinical Syndrome With Germinal Matrix–Intraventricular Hemorrhage








  • Stuttering evolution: hours to days



  • Neurological features



  • Altered level of consciousness



  • Altered motility (usually decreased)



  • Hypotonia



  • Abnormally tight popliteal angle



  • Abnormal eye position or movement or both



  • Respiratory disturbance



Catastrophic Syndrome


The catastrophic syndrome is dramatic in presentation (see Table 24.10 ). The deterioration evolves in minutes to hours and consists of deep stupor or coma, respiratory abnormalities (arrhythmias, hypoventilation, and apnea), generalized tonic seizures, decerebrate posturing, pupils fixed to light, eyes fixed to vestibular stimulation, and flaccid quadriparesis. The clinical distinction between tonic seizures and decerebrate posturing is very difficult in this setting. Indeed, generalized tonic seizures in this setting most often do represent posturing rather than an epileptic phenomenon (see Chapter 12 ). The wide range of frequencies of seizure phenomena recorded with IVH (≈15% to 35%) in part reflects this difficulty. In my experience, seizures occurring with IVH early in the neonatal course usually are associated with periventricular hemorrhagic infarction.


This impressive neurological syndrome is associated with numerous other features—for example, falling hematocrit, bulging anterior fontanelle, hypotension, bradycardia, temperature derangements, metabolic acidosis, and abnormalities of glucose and water homeostasis. The latter include, particularly, inappropriate antidiuretic hormone secretion and, less commonly, diabetes insipidus.


This catastrophic neurological syndrome most likely reflects the movement of blood through the ventricular system, with sequential affection of the diencephalon, midbrain, pons, and medulla. The signs of increased intracranial pressure reflect acute hydrocephalus. The outcome, often poor, reflects the severity of the hemorrhage and, particularly, the extent of complicating parenchymal involvement (see the section on prognosis ).


Saltatory Syndrome


The saltatory syndrome is much more subtle in presentation (see Table 24.11 ). The most common presenting signs are (1) an alteration in the level of consciousness, (2) a change (usually a decrease) in the quantity and quality of spontaneous and elicited motility, (3) hypotonia, and (4) subtle aberrations of eye position and movement (e.g., skew deviation, vertical drift, usually down, and incomplete horizontal movement with the doll’s eyes maneuver). In some patients, disturbances of respiratory function appear to be concomitants, but more data are needed on this issue. In one careful study of serial clinical evaluations and ultrasonographic examinations, an abnormal popliteal angle was found to be a particularly useful diagnostic sign. Eighty-four percent of premature infants with IVH (vs. 10% of infants without hemorrhage) exhibited an abnormally tight angle, perhaps secondary to meningeal irritation. The signs of the saltatory syndrome evolve over many hours, and the deterioration often ceases, only to begin anew after several more hours. This stuttering course may continue for a day or more. The outcome, most often favorable, again relates to the ultimate severity of the hemorrhage and any accompanying parenchymal involvement (see the section on prognosis ).


Clinically Silent Syndrome


The neurological signs of the saltatory syndrome may be so subtle that they are overlooked. Indeed, in a prospective study of infants subjected to clinical assessment and CT scan in the first week of life, only approximately 50% of cases of IVH were correctly predicted to have the lesion on the basis of clinical criteria. The most valuable sign was an unexplained fall in hematocrit or a failure of hematocrit to rise after transfusion. In the serial study of Dubowitz and co-workers, approximately 75% of cases had three or more of the abnormal neurological signs of the saltatory syndrome. Thus, in 25% to 50% of infants with IVH, even careful, serial clinical assessments may fail to reveal a distinct constellation of signs indicative of the lesion.




Diagnosis


Initial Approach


The two essential steps in establishing the diagnosis of IVH are recognition of the clinical setting and use of a suitable screening procedure. In view of the high incidence of the hemorrhage, any very premature infant in a neonatal intensive care facility can be at risk. Thus such infants should be subjected to a suitable screening procedure.


Although the screening procedure of choice is portable cranial ultrasonography (see the section on ultrasound scan later), lumbar puncture (LP) , usually obtained for such purposes as evaluation for meningitis, has provided useful information. The characteristic CSF profile of intracranial hemorrhage consists initially of many red blood cells and elevated protein content, followed shortly by xanthochromia and depressed glucose content (see Chapter 22 ). The first two of these CSF abnormalities are the most critical in early recognition. The degree of elevation of CSF protein correlates approximately with the severity of the hemorrhage. For example, in one study of 48 cases of CT-proven IVH, the mean CSF protein in the small lesions (subependymal hemorrhage or less than 10% of ventricular area filled with blood) was 254 mg/dL; in the moderate lesions (10% to 50% of the ventricular area filled with blood), it was 746 mg/dL; and in the largest lesions (more than 50% of the ventricular area filled with blood), it was 1668 mg/dL. However, the dispersion of the mean values was so large that the CSF protein content could be used only as an approximation of severity of hemorrhage.


In the following sections, the principal means of visualizing germinal matrix hemorrhage–IVH (i.e., ultrasonography, MRI) will be reviewed.


Ultrasound Scan


Ultrasound scan of the neonatal cranium is the procedure of choice in the diagnosis of germinal matrix hemorrhage–IVH. The basic principles of the technique, the features of the instruments used, and the normal anatomical features visualized are described in Chapter 10 . Since the initial reports of the value of the technique in diagnosis of IVH, a vast experience has demonstrated the reliability and versatility of the procedure in this clinical setting. a


a References .

High-resolution imaging, portable instrumentation, lack of ionizing radiation, and relative affordability have been the major advantages. In the following subsections, we illustrate the value of cranial ultrasound scanning in identification of hemorrhage and in determination of timing, severity, and progression of the lesion.


Identification of the Hemorrhage


Ultrasound scan is effective in identification of all degrees of severity of IVH from isolated germinal matrix hemorrhage to major degrees, with or without periventricular hemorrhagic infarction. The physical basis of the dense echoes that correlate with the hemorrhage is probably the formation of fibrin mesh within the clot.


The major elemental lesion, of course, is hemorrhage within the germinal matrix (see Fig. 24.2A ). Intraventricular bleeding results in echogenic material that fills a portion or all of the lateral ventricular system (see Fig. 24.2B and C ). Periventricular hemorrhagic infarction complicating major IVH is a striking echogenic lesion, globular, crescentic, or fan shaped in configuration, usually unilateral, and located on the side of the largest amount of germinal matrix or intraventricular blood or both ( Fig. 24.31 ). The echogenic portion of the lesion is located most commonly in the frontal and parietal regions. The subsequent finding of porencephalic cyst at the site of such a hemorrhagic intracerebral lesion (see Figs. 24.31 and 24.32 ) reflects the essential ischemic nature of the lesion (see the section on mechanisms of brain injury later). The single, large, unilateral, or asymmetrical porencephalic cyst that occurs as a consequence of periventricular hemorrhagic infarction differs from the multiple, small, symmetrical cysts observed as a consequence of periventricular leukomalacia (see Chapter 14 ). The evolution of the typical unilateral or grossly asymmetrical periventricular hemorrhagic infarction after ipsilateral germinal matrix or IVH, or both, has been well documented. Posthemorrhagic ventricular dilation is demonstrated very well by cranial ultrasound scan. This disorder and its management are discussed in detail later (see the section on management ).




Figure 24.31


Ultrasound scans of evolution of periventricular hemorrhagic infarction.

Coronal scans obtained from a premature infant of 30 weeks of gestation on (A–C) day 7 and, D, day 60. The three coronal scans obtained on day 7 were separated by minutes to several hours and show a bulging germinal matrix hemorrhage ( arrow in A) that increases in size (B and C); with the increasing size, a crescentic periventricular echodensity (arrows) consistent with periventricular hemorrhagic infarction develops. Note in C the midline shift to the right. (D) Two months later, a large single porencephalic cyst is observed at the site of the infarction.



Figure 24.32


Periventricular hemorrhagic infarction with evolution to porencephalic cyst, ultrasound scans.

(A) At 9 days of age, the intraparenchymal lesion (p) and ventricular dilation are visible on parasagittal ultrasound scan. Remaining intraventricular clot is also apparent. (B) At 3 weeks of age, the cystic cavity becomes apparent, as necrotic tissue and clot retract. (C) At 2 months of age, a large porencephalic cyst has evolved pari passu with increased ventricular dilation.


Grading the Severity of Hemorrhage


The grading system that we have used is based on the presence and amount of blood in the germinal matrix and lateral ventricles ( Table 24.12 ); determination of the presence of blood in the matrix is best made on the coronal scan, and determination of the amount of blood in the lateral ventricles is best made on the parasagittal scan. In this classification, the presence of periventricular hemorrhagic infarction or of other parenchymal lesions is noted separately because these abnormalities generally are not caused simply by extension of matrix hemorrhage or IVH into normal brain parenchyma (see earlier discussion).



TABLE 24.12

Grading of Severity of Germinal Matrix–Intraventricular Hemorrhage by Ultrasound Scan



















SEVERITY DESCRIPTION
Grade I Germinal matrix hemorrhage with no or minimal intraventricular hemorrhage (<10% of ventricular area on parasagittal view)
Grade II Intraventricular hemorrhage (10%–50% of ventricular area on parasagittal view)
Grade III Intraventricular hemorrhage (>50% of ventricular area on parasagittal view; usually distends lateral ventricle)
Separate notation Periventricular echodensity (location and extent)


Timing of Hemorrhage


Serial ultrasound scans of premature infants have provided invaluable information concerning the time of onset of hemorrhage , and this information, of course, is critical for deciding when to screen for the presence of hemorrhage. In a cumulative series of 105 infants with IVH studied by real-time ultrasonography from the first hours of life, approximately 50% had onset of hemorrhage on the first postnatal day, an additional 25% on the second day, and an additional 15% on the third day ( Table 24.13 ). a


a References .

In a single study of 1105 infants weighing 2000 g or less at birth, approximately 40% of the 265 who developed IVH did so within the first 5 hours of life. The likelihood of onset of hemorrhage on the first postnatal day varies inversely with birthweight; in one series, 62% of hemorrhages in infants between 500 and 700 g birthweight occurred in the first 18 hours. In general, if screening were to be confined to a single postnatal day in the first days of life, a scan on the fourth postnatal day would be expected to detect approximately 90% of all hemorrhages. However, progression of the lesions occurs in approximately 20% to 40% of the affected infants, with maximal extent of the lesion attained usually within 3 to 5 days of the initial diagnosis. a

a References .

Thus a second scan after approximately 5 days is necessary to identify the maximal extent of hemorrhage in the many infants who exhibit progression. I prefer a regimen of two scans in the first week, with timing of subsequent scans determined by the initial findings and clinical events (see later).

TABLE 24.13

Approximate Time of Occurrence of Germinal Matrix–Intraventricular Hemorrhage Identified by Ultrasound Scan



















POSTNATAL DAY PERCENTAGE OF INFANTS WITH GERMINAL MATRIX–INTRAVENTRICULAR HEMORRHAGE a (%)
1 50
2 25
3 15
≥4 10

a Approximately 20%–40% of these infants exhibit progression of hemorrhage over 3–5 days.



Severity of Hemorrhage


The relative distribution of the severity of IVH has been elucidated more effectively with ultrasound scan than was possible with CT scan, because the single CT scan usually obtained could not be expected to identify the maximal severity of hemorrhage in many of the cases. With serial ultrasound scans, this problem is obviated. However, large-scale ultrasonographic studies have used different grading systems and inclusion criteria and have often grouped together infants with grade III IVH and periventricular hemorrhagic infarction. The relative distribution of severity of IVH in infants of less than 1500 g birthweight, based on our unpublished data and that in the literature, is shown in Table 24.14 , Fig. 24.1 . Approximately 20% of the hemorrhages were large (i.e., grade III), with blood usually filling and dilating the lateral ventricles on parasagittal scan (see Table 24.12 for grading system). Approximately 15% of all the infants with hemorrhage had, in addition, large periventricular echodensity consistent with periventricular hemorrhagic infarction. In these infants, the severity of the IVH was grade III in approximately 90%.



TABLE 24.14

Severity of Germinal Matrix–Intraventricular Hemorrhage Identified by Ultrasound Scan



















SEVERITY a PERCENTAGE OF INFANTS WITH GERMINAL MATRIX–INTRAVENTRICULAR HEMORRHAGE (%)
Grade I 40
Grade II 25
Grade III 20
Intraventricular hemorrhage and apparent periventricular hemorrhagic infarction 15 b

From unpublished personal data from approximately 400 premature infants with germinal matrix–intraventricular hemorrhage.

a See Table 24.13 for grading system.


b In approximately 90%, the accompanying intraventricular hemorrhage was grade III in severity.



Particular emphasis should be placed on large IVH with periventricular hemorrhagic infarction , because this lesion accounts for most of the morbidity attributable to IVH per se. This striking lesion is particularly characteristic of the most immature infants (see earlier). Thus, in one series of 2667 infants, IVH with periventricular hemorrhagic infarction accounted for 20% to 30% of all IVH in infants born at 24 to 26 weeks of gestation but less than 5% of all IVH at 30 to 32 weeks of gestation (see Fig. 24.3 ). It is also important to note that the incidence of high-grade IVH (grade III and IV) has not changed markedly overall in the last 20 years in all premature infants less than 28 weeks (see Fig. 24.4 ). However, there has been some decline in infants born at 26 weeks (19% to 11%, P = .03), 27 weeks 15% to 7%, P = .02), and 28 weeks (11% to 5%, P < .01), but not for infants born at 22 to 25 weeks ( Fig. 24.4 ).


Computed Tomography Scan


The CT scan demonstrates the site and extent of IVH very effectively. Indeed, in the first edition of this book, it was stated that “the CT scan is the most definitive means to define the site(s) and extent of periventricular-intraventricular hemorrhage (PIVH).” Ultrasound scan has displaced CT as the principal diagnostic technique, not only because of equivalent resolution for identification of the hemorrhage but also because CT has the disadvantages of requiring the sick premature infant to be transported and of exposing the brain and eyes to ionizing radiation. If MRI is unavailable, CT retains some value, however, for identification of complicating hemorrhagic lesions, such as subdural hemorrhage, hemorrhagic posterior fossa lesions, and certain cerebral parenchymal hemorrhagic abnormalities (see Chapter 10 ).


Magnetic Resonance Imaging Scan


MRI has been shown to provide excellent images of IVH, especially after the first few days of the hemorrhage. However, MRI currently cannot supplant ultrasonography in the evaluation of IVH, because the former technique requires transport to the scanner, has a relatively long data acquisition time, precludes the use of metallic materials still often found on neonatal monitoring and support equipment, and is expensive. The effectiveness of MRI in demonstration of the parenchymal details of periventricular hemorrhagic infarction with germinal matrix hemorrhage–IVH was illustrated earlier (see Figs. 24.12 and 24.14 ).




Prognosis


Prognosis is best considered in terms of the short-term outcome (mortality rate and development of progressive ventricular dilation) and the long-term outcome (neurological sequelae). We will emphasize the relationship of outcome with the severity of hemorrhage and parenchymal abnormalities identifiable on neonatal brain imaging studies—especially the most widely used modality, cranial ultrasonography.


Short-Term Outcome: Mortality Rates and Progressive Ventricular Dilation


The short-term outcome relates clearly to the severity of the hemorrhage and to the degree of prematurity. The mortality rates and incidences of progressive posthemorrhagic ventricular dilation (i.e., hydrocephalus) are shown in Table 24.15 as a function of the severity of the hemorrhage, documented primarily by ultrasound scan, and the infant’s birthweight. Although the data are derived from a single study of 248 infants, findings of other reports are more or less similar. a


a References .

Thus, with small lesions, confined to the germinal matrix or accompanied by small amounts of intraventricular blood (grade I), mortality rates are low, comparable to those of small premature infants without hemorrhage, and the frequency of progressive ventricular dilation in survivors is very uncommon. With moderate (grade II) lesions, mortality rates are higher only in the smallest infants (<750 g), and approximately 5% to 15% of survivors develop progressive ventricular dilation. With severe (grade III) lesions (i.e., blood filling the ventricles), mortality rates are approximately 30% in the infants weighing less than 750 g at birth but still less than 10% in the infants with a birthweight of 751 to 1500 g; approximately 75% of survivors exhibit progressive ventricular dilation in both groups. For those infants who, in addition to severe IVH, also exhibit apparent periventricular hemorrhagic infarction, mortality rates approach 50% in the infants weighing less than 750 g at birth and are approximately 20% in those with a birthweight of 751 to 1500 g, and the incidences of subsequent hydrocephalus are still higher. Indeed, for the now prominent population of infants of less than 750 g birthweight, survival without progressive ventricular dilation is very unusual with these severe lesions. A recent review of changes in a geographical cohort in Nova Scotia revealed that although overall mortality in very preterm infants had significantly decreased over time, from 17.4% during 1993–97 to 7.7% during 2008–10 ( P < .001), mortality in very preterm infants with grade IV IVH (risk 47%) had not changed significantly over time ( P = .152). In addition, the rate of withdrawal of care in IVH was 15.1% for all infants with IVH. This rate had not significantly changed over time ( P = .287). The rate of withdrawal of care in grade IV IVH was 39% for all infants with grade IV IVH and 83% for infants with grade IV IVH who died. These two rates had not significantly changed over time ( P = .475 and P = .275, respectively).

TABLE 24.15

Short-Term Outcome of Germinal Matrix–Intraventricular Hemorrhage as a Function of Severity of Hemorrhage and Birthweight a





































SEVERITY OF HEMORRHAGE b DEATHS IN FIRST 14 DAYS c PVD (SURVIVORS >14 DAYS) c
<750 g ( n = 75) 751–1500 g ( n = 173) <750 g ( n = 56) 751–1500 g ( n = 165)
Grade I 3/24 (12) 0/80 (0) 1/21 (5) 3/80 (4)
Grade II 5/21 (24) 1/44 (2) 1/16 (6) 6/43 (14)
Grade III 6/19 (32) 2/26 (8) 10/13 (77) 18/24 (75)
Grade III and apparent PHI 5/11 (45) 5/23 (22) 5/6 (83) 12/18 (66)

PHI , Periventricular hemorrhagic infarction; PVD , progressive ventricular dilation.

Data from Murphy BP, Inder TE, Rooks V, Taylor GA, et al. Posthemorrhagic ventricular dilatation in the premature infant: natural history and predictors of outcome. Arch Dis Child Fetal Neonatal Ed. 2002;87:F37–F41.

a Values are n (%).


b For grading system, see Table 24.12 .


c Deaths occurring later in the neonatal period are not shown; the total mortality rates (early and late deaths) are approximately 50%–100% greater for each grade of hemorrhage and birthweight than those shown in the table for early deaths alone.



The progressive ventricular dilation that occurs in survivors does not necessarily require a procedure to divert CSF from the lateral ventricles (i.e., ventriculostomy or ventriculoperitoneal shunt). Indeed, many infants, especially with less severe IVH, exhibit cessation of progression, with or without resolution, with no therapy. The natural history of posthemorrhagic ventricular dilation is discussed in more detail in the subsequent section, along with outcomes.


Long-Term Outcome: Neurological Sequelae


The long-term neurodevelopmental outcome of the infant with IVH depends on two key factors: the immaturity of the infant and the degree of parenchymal injury. The latter ranges from germinal matrix destruction to periventricular hemorrhagic infarction (see Fig. 24.9 ). Associated concurrent neuropathologies (see earlier) also may contribute importantly to long-term outcome. There are several limitations to our ability to define the full consequences of IVH in the immature brain, including the inability to assess fully the presence of microstructural brain injury, the impact on subsequent developmental processes, and related factors. Thus, most literature has focused on associations between cranial ultrasound findings and outcome, with only an approximate relationship existing between the quantity of intraventricular blood and the neurologic outcome (see Table 24.14 ). a


a References .

Although the incidence of major neurological sequelae (spastic motor deficits, major cognitive deficits) after minor degrees of hemorrhage is slightly higher than that in infants without hemorrhage and increases to approximately 50% in infants with severe hemorrhage, a clearly higher incidence occurs in infants with IVH complicated by periventricular hemorrhagic infarction or cystic periventricular leukomalacia, or both. Prognostic estimates can thus be refined considerably by assessment of the presence and the degree of parenchymal injury by detailed imaging. It is highly likely that such estimates could also be considerably improved by more sophisticated understanding of the full impact of GMH-IVH on subsequent cerebral development.


A recent meta-analysis summarized nine cohort studies to compare neurodevelopmental outcomes in three groups—no IVH, mild (grade I–II) IVH, and severe (grade III–IV) IVH. (In the analysis, the authors used the term periventricular IVH, and therefore PIVH, rather than GMH-IVH, as in this chapter.) The analysis documented that mild PIVH (two studies, 3508 subjects, unadjusted OR 1.48; 95% CI, 1.26 to 1.73; I 2 = 79%) and severe PIVH (8830 subjects, unadjusted OR 4.72; 95% CI, 4.21 to 5.31; I 2 = 97%) were both associated with higher odds of the primary outcome of death or moderate-severe neurodevelopmental impairment (NDI) at 18 to 24 months of life when compared with infants without IVH. The analysis also evaluated the secondary outcome of moderate-severe NDI, defined as moderate to severe cerebral palsy; moderate to severe cognitive delay; severe visual impairment, defined as visual acuity less than 6/60 (metric scale) in the better eye; or severe hearing impairment, defined as requirement of unilateral/bilateral hearing aids or cochlear implants, distinct from mortality. Mild PIVH was associated with higher odds of moderate-severe NDI compared with no IVH among those who survived to discharge (3032 subjects, unadjusted OR 1.75; 95% CI, 1.40 to 2.20; I 2 = 76%; adjusted OR 1.39; 95% CI, 1.09 to 1.77, I 2 = 70%). Severe IVH was also associated with higher odds of the outcome compared with no IVH (13,691 subjects, unadjusted OR 3.36; 95% CI, 3.06 to 3.68; I 2 = 39%; 2670 subjects, adjusted OR 2.44; 95% CI, 1.73 to 3.42; I 2 = 82%). Finally, severe IVH was associated with higher odds of moderate-severe NDI when compared with mild IVH among survivors (880 subjects, unadjusted OR 2.62; 95% CI, 1.83 to 3.74; I 2 = 0%; 1686 subjects, adjusted OR 2.16; 95% CI, 1.36 to 3.43; I 2 = 0%). This study is informative in two ways: First, and importantly, it highlights the presence of increased risk for NDI in the preterm infant with grade I–II IVH. Second, however, the odds ratio (OR) estimates for each of these outcomes is still relatively low and thus renders individual prognostication challenging. As discussed later, it may be possible to improve prognostications with more advanced neuroimaging techniques.


One of the largest studies contributing to our understanding of the neurodevelopmental consequences of IVH has been the EPIPAGE study that enrolled 1954 infants less than 32 weeks’ gestation and described a clear relationship between greater severity of IVH and an increased risk of adverse neurological outcome. Even in isolated grade I–II IVH the rates of cerebral palsy increased substantially from a baseline of 5.5% to 8.1% for grade I IVH ( n = 229) and 12.2% for grade II IVH ( n = 168). Notably, cerebral palsy rates with isolated grade I–III IVH also rose with immaturity from 5% in infants born at 31 to 32 weeks to 10% to 15% in those born at 27 to 30 weeks and 33% in those born at 24 to 26 weeks. Patra and colleagues also reported that grade I–II IVH was associated with a twofold increase in the risk of lower cognitive performance (Mental Developmental Index) and a 2.6-fold increase in the risk of neuromotor abnormalities (cerebral palsy and tone) after controlling for social and neonatal factors (gender, bronchopulmonary dysplasia, sepsis, necrotizing enterocolitis, maternal marital status, race, and education). Two recent additional studies also showed that grade I–II IVH was associated with worse Mental Developmental Index (MDI) and Psychomotor Developmental Index (PDI) scores, but that this outcome was dependent on gestational age, being significant only at less than 28 to 29 weeks’ gestation. For school-aged outcomes, Sherlock and colleagues examined 298 preterm infants less than 1000 g at age 8 years to determine the impact of IVH in relation to neuromotor and cognitive outcomes. They documented that no IVH was associated with cerebral palsy rates of 6.7%, with no rise in association with grade I IVH (6.4%), but a marked elevation with grade II IVH to 24%.


Despite these large data sets, there continues to be disagreement about the significance of low-grade IVH, with two recent studies having directly conflicting results. Payne and colleagues enrolled 1472 infants, all less than 27 weeks’ gestation. Comparing infants with no IVH and low-grade IVH, they found no difference in rates of cerebral palsy (8% vs. 9%), or NDI (10% vs. 10%), with similarly no increased risk on multivariate analysis. Contrary to this, Bolisetty and colleagues enrolled an alternate cohort also of 1472 infants less than 29 weeks’ gestation and found a consistent association between low grades of IVH and both cerebral palsy (no IVH vs. I–II IVH; 6.5% vs. 10.4%) and moderate-severe neurosensory impairment (no IVH vs. I–II IVH; 12% vs. 22%). On multivariate analysis there remained a one-and-a-half-times increased risk of moderate to severe neurosensory impairment with low-grade IVH, when controlling for gestation, gender, SGA, chronic lung disease, ROP, and PVL. The notable differences between these two studies is that Payne and colleagues had a 1- to 2-week lower mean gestational age for all subgroups, with slightly worse outcomes among infants without IVH, and had fewer infants with a low-grade IVH (270 vs. 336). All of the studies have been limited by the necessity to combine grades I and II IVH into a composite for statistical purposes and by the inability to distinguish the key neuropathologies that may be leading to the subsequent impairment in neurodevelopmental outcome. Cranial ultrasound has been demonstrated to have poor diagnostic utility for diffuse white matter injury, which has been shown to occur in up to 75% of preterm infants (see Chapters 10 and 16 ). Concomitant cerebral white matter injury is also an important determinant of outcome with IVH, as described earlier. Thus low-grade IVH could be a visible marker on cranial ultrasound for the more important neuropathology of cerebral white matter injury, which on MR imaging has been shown to be highly predictive of both motor and cognitive deficits.


Of note, the elevation of risk for poor outcomes with low-grade IVH with increasing immaturity may also suggest an independent role for the loss of the glial and neuronal precursors from the immature germinal matrix. This concept of disturbed cerebral development following low-grade IVH is supported by the work of Vasileiadis and co-workers, who analyzed cerebral volumes on MRI at term equivalent in 12 infants with IVH (seven infants had grade I IVH, four infants had grade II IVH, and one infant had grade III IVH, with no persistent ventriculomegaly; the IVH was bilateral for eight infants). The volumes were compared with 11 preterm infants without IVH. The volume of cortical gray matter on MRI was significantly reduced by 16% in the IVH group (no-IVH group: 122 ± 12.9 mL; IVH group: 102 ± 14.6 mL; F = 13.218). There was no difference in the volumes of subcortical gray matter, white matter, and CSF.


Periventricular Hemorrhagic Infarction—Major Determinant of Long-Term Outcome


Clearly, major determinants of outcome in the premature infant are the presence and severity of associated periventricular hemorrhagic infarction. Although many studies have addressed the outcome in this group, quantitative conclusions are difficult to draw because the selection criteria differ, the numbers of infants are often relatively small, the lesion usually is not quantitated, and the mortality rates vary, in part because of differences in policies of termination of life support in the severely affected infant. Nevertheless, more recent studies a


a References .

provide useful data and complement the largest single study reported earlier ( n = 75). In the latter study, the degree of parenchymal injury was quantitated after identification on ultrasound scan as a large intraparenchymal echodensity (i.e., >1 cm), presumed to represent periventricular hemorrhagic infarction. Among the 75 infants studied, the mortality rate was 59%. This finding should be contrasted with a mortality rate of 8% in the same neonatal unit at the same time for infants with the severest grade of IVH (i.e., grade III IVH but no associated periventricular hemorrhagic infarction). Among the 22 survivors who could be examined on follow-up, 87% exhibited major motor deficits, and 68% had cognitive function less than 80% of normal. The motor deficits correlated with the topography of the parenchymal lesions and thus consisted of either spastic hemiparesis or asymmetrical spastic quadriparesis. In more recent reports, the incidence of major motor deficits has been lower (i.e., ≈50% in 36 surviving infants <32 weeks of gestational age, and 60% in 30 surviving infants <2500 g birthweight ).


Prognostic estimation can be refined by considering the severity of the periventricular hemorrhagic infarction ( Tables 24.16 and 24.17 ). Thus, among infants with extensive lesions (i.e., echodensity that included frontoparieto-occipital regions ( Figs. 24.32 and 24.33 ), 30 of 38 (81%) died, and of the eight survivors, seven had subsequent motor deficits. However, caution is necessary in extrapolating these small numbers to all infants; careful consideration of associated lesions (e.g., periventricular leukomalacia) and other clinical aspects is necessary . Among infants with localized lesions (i.e., echodensity confined to frontal, parietal, temporal, or occipital regions ( Fig. 24.34 ), the outcome was more favorable than after extensive echodensity for both unilateral and bilateral lesions. Thus major spastic motor deficits occurred in only 50% with unilateral localized lesions, and major cognitive deficits appeared in only 12% of these infants. Even with bilateral localized disease, major cognitive deficits occurred in only 50%. In a recent study, only 4 of 12 (33%) infants with unilateral localized lesions had an abnormal motor examination at 2 years of age. Consistent with this more favorable outcome for localized lesions , a study of unilateral periventricular echodensities that evolved to porencephalic cyst reported a developmental quotient less than 80 in five of nine localized lesions, versus six of seven diffuse lesions; spastic motor deficits occurred in four of nine localized lesions versus seven of seven diffuse lesions. The relationship of the superior–inferior involvement of the motor tracts is represented on the cerebral topography of the major motor tracts in Fig. 24.35 . The relationship between the anterior-posterior distribution of periventricular hemorrhagic infarction and outcome is controversial. The discrepancies could relate in part to differing definitions of anterior and posterior. An earlier report indicated that motor deficits were more likely with posterior than anterior lesions, whereas more recent work found more motor deficits with anterior than posterior lesions. Posterior (peritrigonal) lesions have been noted to be especially closely associated with subsequent cognitive deficits and microcephaly.



TABLE 24.16

Long-Term Outcome: Neurological Sequelae in Survivors With Germinal Matrix–Intraventricular Hemorrhage as a Function of Severity of Hemorrhage a



















SEVERITY OF HEMORRHAGE b INCIDENCE OF DEFINITE NEUROLOGICAL SEQUELAE c (%)
Grade I 15
Grade II 25
Grade III 50
Grade III and apparent periventricular hemorrhagic infarction 75

a See text for references. Data are derived from reports published since 2002 and include personal published and unpublished cases.


b For grading system, see Table 24.12 .


c Mean values (to nearest 5%); considerable variability among studies was apparent, especially for the severe lesions. Definite neurological sequelae included principally cerebral palsy or mental retardation, or both.



TABLE 24.17

Outcome of Intraventricular Hemorrhage as a Function of the Severity of Associated Periventricular Intraparenchymal Echodensity






























OUTCOME SEVERITY OF IPE a LOCALIZED
UNILATERAL EXTENSIVE LOCALIZED BILATERAL EXTENSIVE
Mortality rate 21/27 (78%) 8/23 (35%) 9/11 (82%) 6/14 (43%)
Major motor deficits b 5/6 (83%) 4/8 (50%) 2/2 (100%) 5/6 (83%)
Cognitive <75% c 5/6 (83%) 1/8 (12%) 2/2 (100%) 3/6 (50%)

IPE , Intraparenchymal echodensity.

a See text for definitions of “extensive” and “localized.”


b Includes only overt spastic motor deficits.


c Age range at testing generally 1 to 4 years; tests included varying combinations of Bayley, Stanford-Binet, Vineland, Wechsler, and Verbal Language Development Scales.




Figure 24.33


Ultrasound scan of periventricular intraparenchymal echodensity, extensive.

Note that the lesion (arrowheads) extends from frontal to occipital regions.



Figure 24.34


Ultrasound scan of periventricular intraparenchymal echodensity, localized.

Note that the lesion (arrowheads) is confined to the posterior parietal region.



Figure 24.35


Schematic diagram of the location of periventricular hemorrhagic infarction and descending corticospinal tract motor fibers.

Note that the topography of the lesion accounts for the subsequent spastic hemiparesis with prominent involvement of lower as well as upper extremity.


The largest recent series further refine neurodevelopmental outcomes in relation to the regional position of the parenchymal hemorrhage. In the setting of the commonest lesion in the parietal region, cerebral palsy occurred in one-half of infants, with the majority being unilateral spastic in nature. However, an important subgroup of infants with temporal lobe hemorrhages were noted to have a very high risk of behavioral and cognitive challenges ( Fig. 24.36 ).




Figure 24.36


Flowchart of overall review of children with periventricular hemorrhagic infarction (PVHI) treated in a single unit from 1999 to 2012.

Only survivors older than 12 months corrected age were included in the analysis of features of neurodevelopmental outcome. Children with temporal PVHI and bilateral frontal cystic periventricular leukomalacia were excluded. BSCP , Bilateral spastic cerebral palsy; CP , cerebral palsy; USCP , unilateral spastic cerebral palsy.

(From Soltirovska Salamon A, Groenendaal F, van Haastert IC, Rademaker KJ, Benders MJ, Koopman C, de Vries LS. Neuroimaging and neurodevelopmental outcome of preterm infants with a periventricular haemorrhagic infarction. Dev Med Child Neurol . 2014;56:547–555.)




Management


Management of neonatal germinal matrix hemorrhage–IVH is considered best in terms of (1) prevention, (2) initial or acute measures, and (3) treatment of posthemorrhagic ventricular dilation. In the following discussion, we consider these three aspects and conclude with a rational sequence of management.


Prevention


As with many neonatal neurological disorders, the primary goal in management of IVH is prevention. Rational attempts at prevention require an understanding of pathogenesis (see earlier discussion). The relevant prenatal ( Table 24.18 ) and postnatal interventions are discussed next in the context of current concepts of pathogenesis discussed earlier.



TABLE 24.18

Prevention of Germinal Matrix–Intraventricular Hemorrhage: Perinatal Interventions








  • Prevention of preterm birth



  • Transportation in utero



  • Prenatal pharmacological interventions




    • Glucocorticoids



    • Phenobarbital



    • Vitamin K



    • Magnesium sulfate




  • Optimal management of labor and delivery



  • Delayed umbilical cord clamping



  • Umbilical cord milking



  • Temperature stability



  • Newborn resuscitation



Perinatal Interventions


Prevention of Premature Birth.


The most decisive way to prevent IVH would be to prevent premature birth. Those pathogenetic factors referable to the regulation of cerebral blood flow and the microvascular network of the germinal matrix of the premature brain obviously cannot be altered after birth. Indeed, the magnitude of the problem of IVH relates directly to the fact that annually more than 300,000 premature infants (birthweight <2500 g) are born in the United States (≈8% of the 4,000,000 births yearly), and more important, approximately 60,000 of these infants weigh less than 1500 g at birth. Notably, in the United States, the rate of very low birthweight (i.e., <1500 g) infants has actually increased from 1.17% to 1.42% in the past 30 years.


Attempts at prevention of premature birth have been based on three major approaches, operating in sequence: (1) identification of the woman at high risk for premature delivery; (2) management of such a woman with a combination of patient education, treatment of infection, comprehensive health care, and early detection of premature labor; and (3) early treatment of premature labor, primarily with tocolytic agents. Despite comprehensive prevention programs and aggressive use of tocolytic agents, or both, results have not shown consistent benefit. a


a References .



Of particular interest has been the beneficial effect of progesterone or 17-alpha-hydroxyprogesterone caproate (17P) in reduction of preterm delivery. In one large multicenter, randomized clinical trial of 17P begun at 16 to 20 weeks of gestation in women with a history of previous preterm delivery, the rate of preterm delivery in the 17P-treated group was 36% versus 55% in the placebo group. Notably, the incidence of IVH in these infants who weighed less than 2500 g at birth was 1.3% in the 17P-treated group versus 5.2% in the placebo group. A more recent study of 45 singleton pregnancies randomized after successful tocolysis and arrest of preterm labor included 22 women who received 17 P and 23 placebo. The mean gestational age at delivery ( P = .07) and the interval from treatment to delivery ( P = .2) were not affected by 17 P. However, significantly fewer women in the 17 P group delivered at less than 34 weeks (14 vs. 21, P = .03). There was also a significant reduction in the risk of neonatal sepsis ( P = .04) and grade III/IV IVH ( P = .02) in the 17 P group.


Transportation in Utero.


If premature labor and delivery cannot be prevented, then the pregnant woman should be transported to a perinatal center specializing in high-risk deliveries. Infants thus transported in utero have a considerably lower incidence of IVH than apparently similar infants transported after delivery. Whether this difference relates to inherently lower risk in pregnant women who are transported compared with those who are not transported, the type of management of labor and delivery, neonatal resuscitation factors, complications during transport, or a combination of these factors is not yet clear.


Prenatal Pharmacological Interventions.


Because of the possibility that factors related to labor and delivery or to the immediate postnatal period may play a role in the pathogenesis of IVH, a preventive intervention that could be instituted in the presence of impending premature delivery has been sought. Antenatal administration of glucocorticoids , usually betamethasone or dexamethasone, currently is the most clearly beneficial antenatal intervention to decrease the incidence of all varieties of IVH ( Table 24.19 ). The beneficial effect was observed initially in studies of the use of antenatal glucocorticoids to promote fetal lung maturation. An early brief report suggested that this therapy resulted in a decreased incidence of IVH postnatally. In a subsequent careful study, the incidence of germinal matrix hemorrhage–IVH was twofold to threefold lower in infants whose mothers received a complete course of steroids antenatally, compared with infants whose mothers received no steroids or an incomplete course. Similar beneficial effects of glucocorticoid have been well documented. Multivariate analysis suggested that the effect was not related to a lower incidence of complications of respiratory distress syndrome, although the severity of the respiratory disease was less in the treated infants. A large amount of subsequent work demonstrated the beneficial effect of antenatal glucocorticoids ( Fig. 24.37 ). The beneficial effect has been observed most decisively after a complete course of treatment (maternal receipt of two or more doses of glucocorticoid within a week of delivery with an interval of 12 hours from the last dose [or 24 hours from the first dose] to delivery) but also has been observed with a partial course (less than two doses in the week before delivery). a


a References .

Two issues of importance relate to the benefits and hazards of (1) different corticosteroid preparations and (2) repeated courses of antenatal steroids. Concerning the former, betamethasone is preferred over dexamethasone because of more favorable pharmacokinetics, better pulmonary maturation enhancement, and less toxicity. However, some investigators consider the two agents similar in terms of risks and benefits.

TABLE 24.19

Antenatal Steroids and Prevention of Intraventricular Hemorrhage








  • Single most effective antenatal pharmacological intervention for prevention of intraventricular hemorrhage



  • Reduced incidence of both total and severe intraventricular hemorrhage



  • Reduced incidence of cystic periventricular leukomalacia



  • Betamethasone preferred over dexamethasone



  • Mechanism of beneficial effects not established: improved cerebral hemodynamics and maturational benefits most likely




Figure 24.37


Intraventricular hemorrhage and antenatal steroids.

Unadjusted odds ratio and 95% confidence intervals for the occurrence of severe intraventricular hemorrhage after antenatal steroid treatment (vs. no treatment) in three separate studies of infants of less than 1500 g birthweight (the National Institute of Child Health and Human Development Neonatal Research Network [NICHD], the Vermont-Oxford Trials Network [VTOX], and the database of Ross Laboratories [Ross]). The total number of infants was approximately 18,000.

(Data from Wright LL, Horbar JD, Gunkel H, Verter J, et al: Evidence from multicenter networks on the current use and effectiveness of antenatal corticosteroids in low birth weight infants. Am J Obstet Gynecol . 1995;173:263–269.)


Repeated antenatal glucocorticoid courses have not been recommended as routine clinical practice because of adverse effects on fetal growth and cerebral cortical maturation documented in animals and humans. However, one randomized clinical trial ( n = 982) showed benefit of repeat doses for respiratory outcome but no additional benefit concerning incidence of IVH. Another, smaller trial ( n = 249) of mothers who received a course of betamethasone followed by a single booster dose of betamethasone just before preterm birth showed a trend for worse survival and increased respiratory disease but no effect on incidence of IVH for those receiving the booster dose of betamethasone versus placebo.


The most recent Cochrane review of single versus repeated course of antenatal corticosteroids included 10 trials, with a total of 4733 women and 5700 infants. The comparison was between treatment of women who remain at risk of preterm birth seven or more days after an initial course of prenatal corticosteroids with repeat dose(s), compared with no repeat corticosteroid treatment. Repeated corticosteroid reduced the risk of respiratory distress syndrome (RR 0.83; 95% CI, to 0.91). There was no effect on the incidence of IVH. Treatment with repeat dose(s) of corticosteroid was associated with a reduction in mean birthweight (mean difference [MD] −75.79 g; 95% CI, –117.63 to –33.96; nine trials, 5626 infants). At early childhood follow-up, no statistically significant differences were seen for infants exposed to repeat prenatal corticosteroids compared with unexposed infants for the primary outcomes (total deaths; survival free of any disability or major disability; disability; or serious outcome) or in the secondary outcome growth assessments. Finally, twin gestations may not show the same beneficial effects of antenatal corticosteroids, with a recent study showing no difference in respiratory distress syndrome or other neonatal morbidities, including IVH.


The basis for the beneficial effect of antenatal steroids may relate to the improved cardiovascular stability in the treated infants. Thus antenatal steroid therapy is associated with less need for blood pressure support and less hypotension postnatally. That this beneficial postnatal hemodynamic effect could be related to improved placental blood flow (and thereby less likelihood of impairment of the infant’s cerebrovascular autoregulation) seems possible because antenatal betamethasone has been shown to lead to a decrease in placental vascular resistance. Perhaps relevant in this context is the observation that antenatal steroid administration is associated with a reduced incidence of cystic periventricular leukomalacia postnatally. The possibility also exists that the therapy leads to the beneficial effect on IVH in part by stimulation of maturation of brain structures (e.g., germinal matrix).


In conclusion, antenatal corticosteroids reduce the risk for IVH in singleton preterm infants, particularly when a completed course is given and betamethasone is the agent administered. The biological basis for this benefit remains unclear. Optimal preparations, dosing, and timing to delivery remain worthy of investigation, particularly in the setting of multiple births.


Antenatal administration of phenobarbital led to interesting results in five studies. Initial studies raised the possibility that antenatal phenobarbital may have a small protective effect against IVH. However, later work failed to show a significant protective effect. Currently this approach appears not to be beneficial for the prevention of IVH.


Because the function of vitamin K–dependent coagulation factors in preterm infants is approximately 30% to 60% of the function in adults, vitamin K was administered intramuscularly to women in premature labor at least 4 hours before delivery in an attempt to prevent IVH. The incidence of IVH in the prenatally treated infants was 5%, compared with 33% in the control infants. Although the infants treated prenatally with vitamin K had normal prothrombin activity (compared with 67% of normal in control infants), no statistically significant relationship existed between prothrombin activity and the occurrence of IVH. In a subsequent larger study ( n = 100), antenatal administration of vitamin K resulted in a lower incidence of total IVH (16% vs. 36% in control infants) and of severe IVH (0% vs. 11% in control infants). However, uncertainty concerning the role of vitamin K per se persists, despite these interesting data. Moreover, two later studies of antenatal vitamin K administration, combined in one of the studies with antenatal phenobarbital administration, showed no significant beneficial effect on hemostasis or incidence and severity of IVH. Thus, on balance, it does not appear that antenatal vitamin K is useful for the prevention of IVH.


As described in Chapter 16 , some data, although not consistent, indicate that the use of antenatal magnesium sulfate (principally for tocolysis) is followed by a lower incidence of cerebral palsy in the premature infants so treated. Although one preliminary report suggested that antenatal magnesium sulfate therapy results in a lower incidence of “grade III or IV IVH,” most data do not show a beneficial effect on IVH or cerebral palsy, or both. More concerning, some reports noted an increase in perinatal and postnatal mortality after antenatal magnesium administration. Other studies did not show this increased mortality. A more recent report showed a decreased incidence of IVH after the combination of antenatal magnesium and aminophylline, but the population was not randomized, and the numbers of infants with IVH were small (1 of 78 in the treated group and 7 of 68 in the control group). A review of the nine best trials concluded that antenatal magnesium sulfate has no beneficial effect on “the risk of neonatal morbidity.”


Optimal Management of Labor and Delivery—Delivery Mode.


As discussed earlier concerning pathogenesis, potentially deleterious effects of labor and delivery relate principally to the easily deformed, particularly compliant skull of the premature infant. Such deformations presumably could lead to dangerous elevations of venous pressure and perhaps to an impairment of cerebrovascular autoregulation (analogous to that observed in adult patients with head trauma). Prolonged labor and breech delivery would be considered most likely to lead to such hemodynamic effects, and some, but not all, work supports this contention. a


a References .

In one large prospective study that used multivariate analysis (see Table 24.7 ), abdominal delivery appeared to be protective concerning germinal matrix hemorrhage, and longer duration of labor was deleterious. In a later study, cesarean section before the active phase of labor resulted in a lower frequency of progression to severe IVH but did not affect total incidence of IVH. Another study identified a twofold lower incidence of grade III/IV IVH in infants delivered by cesarean section without labor versus section delivery with labor, but the difference disappeared after logistic regression analysis. Thus the potential value for cesarean section in selected preterm infants for the prevention of IVH is suggested by some, but not all, work; more data are needed to define the specific clinical circumstances that should lead to a recommendation for abdominal delivery.


Delayed Cord Clamping.


DCC by 30 to 120 seconds rather than immediate clamping has been reported to be associated with less need for transfusion and lower rate of IVH. In December 2012, the American College of Obstetricians and Gynecologists recommended a delay of 30 to 60 seconds in umbilical cord clamping for all preterm deliveries. Most recently in January 2017, the American College of Obstetricians and Gynecologists recommended a delay in umbilical cord clamping in vigorous term and preterm infants who do not require resuscitation for at least 30 to 60 seconds after birth.


Although DCC has been associated with a decrease in the overall incidence of IVH, it has not been shown to have a beneficial effect on severe IVH, and thus its clinical application has not been uniformly embraced. It is also worthy of note that both the ACOG and ILCOR recommendations are in vigorous preterm infants. There are no data on DCC in the premature infant in need of resuscitation.


With regard to subsequent outcomes, a recent randomized controlled trial reported the effects of placental transfusion on neonatal and 18-month outcomes in preterm infants with singleton fetuses ( n = 208) between 24 weeks of gestation and 32 weeks of gestation. There were no differences in the number of infants who received phototherapy, days of phototherapy, delivery room resuscitation, Neonatal Acute Physiology scores at 12 hours of age, and the rates of IVH or late-onset sepsis between the groups. However, at 18 to 22 months, DCC was found to be protective against motor scores less than 85 on the Bayley Scales of Infant Development. The authors concluded that although DCC did not alter the incidence of IVH or late-onset sepsis in preterm infants, it improved motor function at 18 to 22 months’ corrected age.


Umbilical Cord Milking.


An alternative to DCC is umbilical cord milking (UCM), a procedure that can be performed in 20 seconds. UCM is performed by holding the infant at or 20 cm below the level of the placenta. The cord is pinched as close to the placenta as possible and milked toward the infant for 2 seconds. The cord is then released and allowed to refill with blood for 1 to 2 seconds between each milking motion, a process repeated a total of 4 times. After completion, the cord is clamped and the newborn handed to the resuscitation team. Milking the cord 4 times provides a similar amount of placental-fetal blood as with DCC for 30 seconds.


A recent meta-analysis of UCM, including seven randomized clinical trials involving 501 infants delivered at less than 33 weeks, demonstrated that infants who underwent UCM had a higher hemoglobin level and lower risk of oxygen requirement at 36 weeks and IVH of all grades compared with those who underwent immediate cord clamping. A recent pilot study of 75 extremely premature neonates (born at a gestational age <29 weeks) randomly assigned to receive UCM or immediate cord clamping also demonstrated a 50% reduction in total IVH with UCM. Moreover, in another recent retrospective study of 318 infants born at less than 30 weeks, UCM was associated with reductions in IVH, necrotizing enterocolitis, and death before hospital discharge. Concerning the mechanism of benefit from UCM, a further study that enrolled 197 infants (mean gestational age 28 ± 2 weeks) reported a higher hemoglobin level at birth, improved hemodynamics (higher blood flow and improved blood pressure), and improved urine output with UCM compared with DCC in preterm infants delivered by cesarean section. The authors also noted SVC flow and higher right ventricular output in infants treated with UCM.


A concern has been raised over UCM relating to the rapidity of the delivery of the bolus of blood. Rapid changes in venous pressure during UCM were addressed in an early trial that demonstrated no greater increase in venous pressures with UCM compared with uterine contractions or a newborn cry during intact placental circulation. A recent meta-analysis evaluating the safety and efficacy of UCM at birth concluded that there was a lower risk of oxygen requirement at 36 weeks and IVH of all grades.


The potential for developing hyperbilirubinemia is another issue of concern with UCM. A Cochrane review found that none of the infants with elevated bilirubin levels required phototherapy treatment or exchange transfusions. However, in populations more susceptible to neonatal hyperbilirubinemia, especially those of Asian ethnicity, careful monitoring of the serum bilirubin level is warranted.


According to the 2010 International Consensus on Cardiopulmonary Resuscitation and Emergency Cardiovascular Care Science With Treatment Recommendations and subsequent review of both DCC and cord milking in preterm newborns in the 2015 Umbilical Cord Management in the International Liaison Committee on Resuscitation (ILCOR) systematic review, DCC for longer than 30 seconds was proposed as reasonable for both term and preterm infants who do not require resuscitation at birth. However, it was felt that there was insufficient evidence to recommend “delayed” cord clamping for infants who required resuscitation at birth, and more randomized trials involving such infants were needed. ILCOR also suggested that cord milking should not be routinely used for infants born at less than 29 weeks of gestation outside of a research setting because of concerns over rapid volume changes. Further studies are warranted to elucidate this issue, because cord milking may improve the initial mean blood pressure, hematologic indices, and reduce intracranial hemorrhage. However, there is currently no evidence with regard to improvements in long-term outcomes.


Temperature Stabilization.


Unless significant preventative efforts are made, periviable newborns will quickly lose heat in the delivery room by evaporation of amniotic fluid from the baby’s body, by conduction of heat from the body touching cooler surfaces, by convection to cooler surrounding air, and by radiation to cooler objects in the vicinity. For every 1°C below 36°C on admission temperature, mortality increases by 28%. The periviable infant should be placed in a high diathermancy food-grade polyethylene bag or wrap without initial drying up to the level of the shoulders. Multiple studies have shown that plastic bags and wraps improve temperature on admission to the NICU, but as of yet there are no studies powered for important clinical outcomes, such as mortality or IVH.


Although normothermia may reduce cold stress , there are few studies to support an association of admission temperature or admission hypothermia to an increased risk for IVH. A single observational study of 271 VLBW infants showed that after correction for confounders by multivariate logistic regression analysis admission hypothermia (at ≤35.5 and at ≤35°C) was not associated with IVH.


Newborn Resuscitation.


Consideration of the intravascular pathogenetic factors makes it clear that certain practices in newborn resuscitation may increase the likelihood of IVH in the premature infant ( Table 24.20 ). In particular, overly rapid infusion of volume expanders or of hypertonic solutions, such as sodium bicarbonate, should be avoided.



TABLE 24.20

Progressive Ventricular Dilation After Intraventricular Hemorrhage








  • Etiology: acute, particulate blood clot; chronic, obliterative arachnoiditis in posterior fossa; aqueductal obstruction less commonly



  • Temporal features: usual onset of progression 1–3 weeks after hemorrhage; rapidity of evolution directly related to severity of hemorrhage



  • Rapid head growth or signs of increased intracranial pressure or both following ventricular dilation by days to weeks



  • Posterior horns of lateral ventricles dilating before, and more severely than, anterior horns



Concerning cerebrovascular autoregulation, the most important admonition in neonatal resuscitation is to establish adequate ventilation promptly to prevent hypoxemia and hypercarbia, two alterations that result readily in pressure-passive cerebral circulation. Because of the latter facts and because hyperventilation in animals and humans sufficient to decrease Pa co 2 to approximately 25 mm Hg restores autoregulation after hypoxia, two retrospective studies of hyperventilation in the first 2 hours of life and the subsequent occurrence of IVH were conducted. The data in the initial study suggested that Pa co 2 values less than 35 mm Hg led to a decrease in incidence of subsequent IVH, but a subsequent study failed to confirm this observation. Currently, it seems prudent to recommend that adequate ventilation be established promptly in the resuscitation of the newborn infant, and that hypoxemia and hypercarbia be avoided. However, this goal does not require systematic early intubation of all ELBW infants (≤1000 g) at the slightest signs of respiratory distress. An individualized approach to intubation is important. It is also important that if continuous positive airway pressure (CPAP) is to be used as the principal mode of ventilator support, an adequate but not excessive end expiratory pressure must be provided, with the goal of avoiding overdistention or elevated intrathoracic pressures.


Postnatal Interventions


Postnatal interventions aimed at reducing the risk for IVH have principally focused on cardiorespiratory management to reduce fluctuations in cerebral perfusion and pharmacological approaches to enhance cerebral blood flow and improve vascular stability.


Correction of Fluctuating Cerebral Blood Flow Velocity.


The nearly invariable relationship between fluctuating cerebral blood flow velocity in the ventilated premature infant with respiratory distress syndrome and the subsequent occurrence of IVH (see Table 24.5 ) led to a search for interventions that could prevent this hemodynamic disturbance. Muscle paralysis with pancuronium bromide was found to be highly effective for the rapid conversion of the fluctuating pattern to a stable velocity ( Fig. 24.38 ). Moreover, muscle paralysis eliminated the fluctuations in venous pressure that accompany the cerebral arterial fluctuations (see earlier discussion). Thus a prospective, randomized study of muscle paralysis from the first day of life to 72 hours of age in ventilated premature infants with the fluctuating hemodynamic disturbance was undertaken. All the control infants (i.e., nonparalyzed) developed IVH, consistent with previous observations. After completion of the controlled study, a second study in 72 ventilated premature infants with the fluctuating circulatory abnormality for the first 72 hours of life showed a prominent reduction in IVH, including especially severe IVH.




Figure 24.38


Effect of muscle paralysis on fluctuating cerebral blood flow velocity from a ventilated, preterm infant with respiratory distress syndrome.

Before paralysis, the constantly fluctuating peak systolic and end-diastolic flow velocities are apparent. After paralysis was induced by pancuronium, the fluctuating pattern was eliminated.


Importantly, the fluctuations were related to the infant breathing out of synchrony with the ventilator, inducing a pulses paradoxus –like effect. Paralysis eliminated the infant’s own breathing activity with immediate stabilization of the blood pressure and fluctuating cerebral blood flow velocity patterns. Over the last 10 years, several clinical and therapeutic factors have reduced the occurrence of this potential deleterious relationship. First, the near-universal use of antenatal glucocorticoids has reduced the severity of respiratory distress syndrome and improved the stability of the infant’s blood pressure. Consequently, there has been a lesser requirement for volume expanders and/or inotrope use. Second, the early use of surfactant replacement therapy for infants with evidence of respiratory distress syndrome has facilitated the earlier extubation of infants, reducing the severity of their respiratory instability and the length of time that the infant is ventilated. Third, modern ventilators now have sophisticated feedback strategies that facilitate the infant’s breathing in synchrony with the ventilator. In this regard, in a report in 2015 of 47,816 VLBW infants in the Vermont Oxford Network, only 50% of these infants ever received any positive pressure ventilation. Sixty percent of infants were ventilated for less than 24 hours, and 80% for less than 7 days. Thus it is the most immature and sick infants (<20% of VLBW infants) who may remain ventilator dependent during the first few critical days when the risk for IVH is highest and reducing fluctuations in systemic and cerebral perfusion are most often considered.


For those infants who may appear irritable , the use of sedatives is a clinical option. In our own experience and that of the more recent published literature, the need for muscle paralysis now is rare. Of note, however, the use of opiates and benzodiazepines has increased dramatically over the past two decades in some centers ( Fig. 24.39 ). The factors responsible for this increase in use are unclear, but it has not been driven by clear data indicating that such practice reduces the risk for IVH.




Figure 24.39


Opioid and benzodiazepine administration compared with ampicillin and inotrope administration over time.

(From Zimmerman KO, Smith PB, Benjamin DK, Laughon M, Clark R, Traube C, Sturmer T. Hornik CP. Sedation, analgesia, and paralysis during mechanical ventilation of preterm infants. J Pediatr . 2017;180:99–104, figure 2.)


In view of the increased use of sedation rather than paralysis, it is plausible that the use of such sedatives may reduce the tendency for fluctuating cerebral blood flow associated with handling, painful procedures, and/or mechanical fighting of the ventilator, thereby leading to lower rates of IVH. Nevertheless, the impact of opioids on acute brain injury and long-term neurodevelopment in mechanically ventilated preterm neonates has been examined in two large randomized controlled trials, where no difference in the composite outcome of severe IVH, PVL, or death was detected. It is worthy of note that infants that required additional boluses of morphine in the morphine group demonstrated an increased risk for IVH. However, long-term follow-up was also concerning, as it revealed conflicting results regarding the neurodevelopmental impact of early morphine exposure in preterm neonates. Even though the randomized Neurologic Outcomes and Preemptive Analgesia in Neonates study (NEOPAIN) showed no difference in overall intelligence between groups, morphine exposure was associated with a smaller head circumference, impaired short-term memory, and social problems at 5 to 7 years of age compared with the placebo group. An understanding of how to best target sedation and/or paralysis is limited in these studies because of a lack of knowledge of cerebral perfusion and autoregulation. If accurate monitoring of cerebral perfusion were achievable, then a more rational targeted approach would identify which infant might benefit from sedation/paralysis to minimize any fluctuations in cerebral blood flow.


Correction or Prevention of Other Major Hemodynamic Disturbances.


As indicated in the earlier discussion of pathogenesis, increases and decreases in cerebral arterial blood flow and increases in cerebral venous pressure can be involved in the pathogenesis of IVH. Thus care must be taken to prevent sharp elevations in blood pressure and cerebral blood flow with excessive handling, tracheal suctioning, rapid infusions of blood or other colloid, exchange transfusions, apneic spells, seizures, pneumothorax, and hypercarbia. As noted earlier, the use of antenatal corticosteroids, coupled with targeted surfactant replacement therapy, has reduced many of these events, particularly pneumothorax. Fluctuating Pa co 2 or hypocarbia also should be avoided. Several smaller retrospective studies suggested that alterations in various monitoring and caretaking procedures, resulting in minimal stimulation , led to a decrease in incidence of IVH. However, out of necessity, the use of historic controls made it difficult to specify the most important alterations. A subsequent prospective study in which infants were randomly assigned to a carefully quantitated reduced manipulation protocol ( n = 62) or to standard care ( n = 94) did not show a significant difference in incidence of IVH (30% in study infants, 37% in control infants).


In an attempt to avoid (rapid) fluctuations in CBF during routine care, several nursing interventions have been proposed. These nursing interventions are especially important during the first 72 hours after birth because most GMH-IVH develops during this time frame. The first of these interventions consists of positioning the head of the infant in a neutral (i.e., midline) position, enabling optimal cerebral venous drainage through the internal jugular veins. As a consequence, hampered venous drainage and ultimately bleeding is believed to be prevented. Low-quality evidence obtained in very small studies indicates that there is no significant effect on cerebral oxygenation by head rotation and/or head tilting in preterm infants. One study performed by Liao and co-workers revealed a small, statistically significant, one-sided decrease in rScO 2 after head rotation to the left. However, this decline of only 1% is unlikely to be of clinical significance. The second proposed intervention consists of elevating the head of the incubator 15 to 30 degrees upward (i.e., tilting) to facilitate venous outflow from the brain by promoting hydrostatic cerebral venous drainage.


Finally, elevations in venous pressure must be avoided by prompt treatment of myocardial impairment (e.g., in the asphyxiated infant) and factors that may increase intrathoracic pressure and thereby cerebral venous pressure, such as pneumothorax and vigorous tracheal suctioning. A more extensive evaluation of myocardial function by functional echocardiography may assist in the decision to reduce cardiovascular afterload or manage preload. Management that is limited to evaluation of a single mean arterial blood pressure will likely result in inadequate or inappropriate therapy.


The role of surfactant per se in prevention of the hemodynamic disturbances associated with mechanical ventilation of infants with respiratory distress syndrome and thereby IVH is not entirely clear. Thus a series of earlier studies using one of at least seven different preparations of surfactant in prophylactic or rescue trials resulted in findings that were not entirely uniform. In general, however, neonatal mortality rates, severity of respiratory distress, and air-block complications (e.g., pneumothorax) were reduced. The incidence and severity of IVH most often were either unchanged or reduced. The occurrence, albeit unusual, of increased incidence of IVH in surfactant-treated infants and the failure of a consistent decrease in incidence of IVH (despite a decrease in respiratory disease) in such infants led to the suspicion that surfactant treatment may have deleterious cerebral hemodynamic effects. Available data suggest that surfactant therapy may cause a transient increase in cerebral blood flow velocity and cerebral blood volume and electroencephalographic depression, but the effects are generally not marked. Moreover, no clinical or biochemical evidence of deleterious effects (e.g., serum CK-BB levels) has been demonstrated. In addition, the combined use of antenatal glucocorticoids and postnatal surfactant has added benefits concerning the prevention of IVH.


Alternative methods of mechanical ventilation (i.e., high-frequency oscillatory ventilation or high-frequency jet ventilation) are not clearly superior to conventional ventilation regarding the incidence of IVH. Indeed, in initial work, the incidence of severe IVH was higher in infants treated with high-frequency oscillatory ventilation (26%) than in infants treated with conventional mechanical ventilation (18%), and the neurological outcome was poorer in the former group of infants. A subsequent multicenter study showed a similarly higher incidence of IVH (36% vs. 20%), but four later studies showed no increase in incidence of IVH in infants treated with high-frequency oscillatory ventilation. A significantly deleterious effect concerning IVH also was not observed in two studies of high-frequency jet ventilation, although an increase in the incidence of cystic periventricular leukomalacia was documented in one report. More data are needed on these issues. Most frequently, early nasal CPAP is being used in the delivery room or early in the neonatal course. A meta-analysis of 2364 infants comparing CPAP with mechanical ventilation, CPAP resulted in a small but clinically significant reduction in the incidence of BPD at 36 weeks (typical RR 0.89; 95% CI, 0.79 to 0.99) and death or BPD (typical RR 0.89; 95% CI, 0.81 to 0.97). There was no difference in the rates of IVH. Early CPAP did appear to be associated with a higher risk for pneumothorax if surfactant therapy was not administered.


The use of inhaled nitric oxide for premature infants with severe respiratory failure, related most often to severe respiratory distress syndrome, currently is under active investigation. The gas is used for pulmonary benefits related to its strong pulmonary vasodilator properties. Thus far, no consistent significant differences in the rate of IVH in infants treated with nitric oxide have been reported. One controlled study of 793 infants 34 weeks of gestational age or younger described a decline in grade 3 or 4 hemorrhage in infants treated with nitric oxide from the first days of life but only in infants of birthweight 750 to 999 g. Another report described improved neurodevelopmental outcome in infants treated with inhaled nitric oxide, but the neuropathological basis for the improvement was not clear.


Correction of Abnormalities of Coagulation.


Although in selected infants abnormalities of coagulation (or of platelet-capillary interactions) may play a role in the pathogenesis of IVH, it is unclear whether interventions to correct or prevent such abnormalities are indicated for all premature infants. Thus in one controlled study of the administration of fresh frozen plasma (10 mL/kg) to premature infants on admission to the nursery and again at 24 hours of life, treated infants exhibited a decrease in the overall incidence of IVH (14% vs. 41% in untreated infants). However, no difference in incidence of severe IVH was noted, and no clear effect on coagulation variables could be demonstrated. The possibility was raised that the fresh frozen plasma exerted its benefit by “stabilizing the circulation” rather than by an effect on coagulation. Two later studies of administration of fresh frozen plasma showed no benefit regarding prevention or extension of IVH or on neurological outcome at 2 years. Thus, at present, no clear indication exists for the routine administration of fresh frozen plasma postnatally to premature infants for prevention of IVH.


Pharmacological Interventions


Indomethacin.


In the last edition of this book, we reviewed 19 controlled studies of indomethacin as a means to prevent IVH. The drug was studied particularly for prophylaxis, rather than treatment, of patent ductus arteriosus, and it was generally administered initially before 12 hours of age. A summary of the pooled results showed that indomethacin administration led to a decrease in the incidence of overall IVH (RR, 0.88; 95% CI, 0.80 to 0.96) and of severe IVH (RR, 0.66; 95% CI, 0.53 to 0.82). In the largest early series ( n = 431), Ment and co-workers observed a decrease in the incidence of total IVH (18% to 12%) and grade IV IVH (4.5% to 0.5%). However, in this study, early-onset IVH was excluded, and the marked preponderance of grade IV IVH relative to grade III IVH (10 : 1) in the control population was unusual and raised the possibility of an undefined unusual characteristic of the control population. Indeed, in centers where the incidence of grades III and IV IVH in the control population was greater than 10%, indomethacin administration was shown to lead to a significant reduction in these severe hemorrhages (OR, 0.54; 95% CI, 0.36 to 0.82; P < .005), whereas in centers where the incidence was less than 10%, indomethacin did not lead to a significant reduction (OR, 0.72; 95% CI, 0.39 to 1.33; P < .3).


An interesting development in this area was the repeat analysis of the data from the earlier study of Ment and co-workers ( n = 431; Table 24.21 ). Thus the beneficial effect of indomethacin on IVH was shown in male but not female patients. A repeat analysis of the later larger study of prophylactic indomethacin ( n = 1202) showed a weak differential effect of indomethacin by sex; for severe IVH, the incidences in indomethacin-treated versus placebo groups were 9.8% versus 11.7% (OR, 0.46 to 1.44) for female patients but 8.6% versus 14.7% (OR, 0.31 to 0.94) for male patients. Thus, on balance, in the total experience with indomethacin, a generally favorable preventive effect of the drug on IVH seems apparent, particularly or exclusively in male patients . Follow-up of the large series of infants studied by Ment and co-workers showed no difference in the development of ultrasonographically demonstrated cystic periventricular leukomalacia or in incidences of cerebral palsy or of cognitive impairment at 36 months of age. Other investigators also have shown no beneficial long-term neurodevelopmental effects. However, again, when male and female patients are analyzed separately, a clear cognitive benefit is apparent in male but not female patients ( Table 24.22 ).


May 16, 2019 | Posted by in NEUROLOGY | Comments Off on Preterm Intraventricular Hemorrhage/Posthemorrhagic Hydrocephalus

Full access? Get Clinical Tree

Get Clinical Tree app for offline access