1.gif (1892 bytes)

Review Article

Indian Pediatrics 1998; 35:1081-1096 

Surfactant Replacement Therapy


Victor Yu
 

Reprint requests: Dr. Victor Yu,.Professor of Neona- tology, Monash University, Director of Neonatal Intensive Care, Monash Medical Center, 246, Clayton Road, Clayton Victoria 3168, Australia.
Email: [email protected]

Prevention of respiratory distress syndrome (RDS) is best achieved with prevention or effective treatment of preterm labor, failing which antenatal corticosteroid therapy is the next most efficacious intervention. Postnatal prevention of RDS is the remaining option after the above measures have been taken, and must not be solely relied upon in the place of good obstetric practice. Postnatal prevention of RDS depends on general measures such as prompt treatment of perinatal asphyxia and a high quality of neonatal care. This review focuses' on the use of surfactant therapy as an important component of clinical practice within the neonatal intensive care unit for the management of RDS, in particular its successes and remaining controversies, and surfactant therapy for other indications.

Types of Surfactant

Studies published in the 1970s showed that natural surfactant derived from animal lungs improved lung function and pre- vented respiratory distress in prematurely delivered animals. In 1980, the first successful human study was published, showing that surfactant replacement therapy improved oxygenation, ventilatory requirements, X-ray abnormalities, acidosis and hypotension in.1O pre term infants with RDS(1). Surfactants are of two general classes, natural and synthetic surfactants. Natural surfactants are either heterologous (from minced bovine lung tissue, e.g., Survanta, or extracted from saline lavage fluid of calf lungs, e.g., Infasurf, or from porcine sources, e.g., Curosurf) or homologous (human aminiotic fluid from term pregnancies). Some are. modified by addition of dipalmitoyl phosphotidylcholine, palmitic acid and triglyceride (Survanta) or by removal of neutral lipids by chromato- graphy (Curosurf) to improve the surface properties of the extract. Synthetic surfactants (e.g., Exosurf) contain dipalmitoyl phosphotidylcholine,the main surface- active phospholipid of surfactant, and other components which facilitate surface adsorption. Natural surfactants contain hydrophobic surfactant proteins (SP) Band C which aid in surfactant adsorption and resist surfactant inactivation(2). The main structural properties of these proteins have been clarified which makes it possible to '. design synthetic analogues for use in synthetic surfactants(3). A synthetic peptide (KU) which mimics the aminoacid pattern of SP-B has been found to be effective in treatment of infants with RDS(4).

Clinical Effects of Surfactant Therapy

Since the first randomized clinical trial (RCT) was published on surfactant replacement therapy in 1984, over 50 such trials have been reported. These RCTs have set a new standard for the evaluation of new therapeutic innovations in neonatology. Meta-analyses based on these RCTs have been published(5,6). These showed that by decreasing the need for oxygen and ventilatory support, pneumothorax (PTX) and 1081 mortality rates were reduced. The odds ratio for neonatal mortality was about 0.6; a 30-40% reduction in mortality is expected with surfactant therapy compared to controls. The efficacy of surfactant therapy demonstrated in RCTs has been shown to be translated into effectiveness in routine clinical use. Two observational studies, each involving over 4000-5000 infants weighing < 1500 g in 8 and 14 US centers respectively over a 3-5 year period, reported a 30% reduction in mortality after surfactant was introduced(7,8). These studies estimated that 50-80% of the largest drop in national infant mortality observed in 20 years in the USA (from 9.7 to 9.1 per 1000 births between 1989 and 1990) could be attributed solely to the introduction of surfactant therapy.

Surfactant therapy, by improving cardiorespiratory stability and oxygenation, should theoretically reduce non-pulmonary complications of prematurity such as intra- ventricular hemorrhage (IVH), necrotising enterocolitis and retinopathy of prematurity (RaP). However, published clinical trials have not consistently reported such benefits. Several trials did find a significant improvement in the rate of IVH(9,10) but a multicentre trial in Europe showed an increased risk of periventricular leukoma-Iacia(11). Electroencephalographic data showing periods of electrical silence lasting up to 20 minutes after administration of surfactant was thought, to be suggestive of cerebral ischemia(12) though this was found not to be directly related to alterations in blood gases or systemic circulation(13). A reduction in cerebral blood flow velocity (CBFV) after surfactant administration was interpreted as resulting from a fall in cerebral blood flow(14). A literature review suggested an overall increased risk' for germinal matrix and small IVH with surfactant therapy in infants of 600-750 g but no increased risk, for large IVH or intraparenchymal hemorrhage(15).

It concluded that the risk of IVH was related to the management of surfactant delivery. A learning curve operates in the use of surfactant in regards to anticipatory ventilator management to minimize cardio-respiratory instability following surfactant administration. With careful management of oxygenation,1 and ventilation as surfactant therapy becomes more familiar and customary  only small and transient perturbations occur with cerebral oxyhemoglobin concentration and cerebral blood volume measured by near infrared spectro-scopy(16).

Although a greater proportion of preterm infants are surviving with surfactant therapy, the risk of ROP among the survivors was lower than control infants(17-19). There was also no difference in their prevalence of neurodevelopmental disability, growth retardation and late respiratory or allergic disease(20-39). Indeed, 9 of the 10 double-blind RCT follow-up studies published in 1995(30-39) reported lower proportions and absolute numbers of infants with disability with significant gains in developmental stores in the surfactant-treated group. No adverse long- term effect on pulinonary function was identified as the two groups were found to have similar functional' residual capacity (FRC), tidal volume, compliance and time constant of the respiratory system(26-27). One study showed that surfactant treatment was associated with beneficial long-term effects on resistive airflow properties as reflected by a lower resistive work of breathing, improved expiratory reserves and reduced evidence of airflow obstruction(28).

Surfactant therapy has become routine in neonatal intensive care units except in some developing countries. A number of questions which have been raised on how to optimize its use. These include the method of administration, timing of therapy, amount to give and number of doses, type of surfactant, reason for non- responsiveness, cost implications, role of adjunctive therapies, and surfactant for indications other than RDS.

Method of Administration

It is generally recommended that the surfactant is delivered as a bolus directly into the lungs through an endotracheal tube over a IS-minute period, either via a catehter inserted into the endotracheal tube in 2-4 aliquots after disconnecting the infant from the ventilator, or into the side-port on the endotracheal tube adaptor without the need of removing the infant from the ventilator. Alternatively, surfactant can be delivered via a catheter inserted into the endotracheal tube via a side-port in the ventilator circuit adjacent to the endo-tracheal adaptor without. the need of removing the infant from the ventilator. Infants who remained connected to the ventilator during surfactant instillation have been shown in a RCT to experience less oxygen desaturation compared to those who were disconnected(40). The effect of maintaining a positive end-expiratory pressure during surfactant instillation has also been shown to result in more homogeneous distribution of surfactant within the lungs(41).

Several protocols have been advocated to vary the position of the infants during administration to enable uniform distribution of sufactant to the alveoli, but their efficacy has not been tested. Different protocols of positioning were found in a RCT not to affect the infants' clinical outcomes(40), and a study using an animal model showed that keeping the chest in the horizontal may result in the most even distribution of the surfactant in the two lungs(42). This simple practice may eliminate possible hypoxemia and cardiovascular disturbances associated with the handling of the infants using current positioning techniques. Rapid instillation over S minutes is not recommended as it results in an increase in CBFV and pCOz and requires closer monitoring of blood gases to maintain adequate ventilation compared to a slower IS-minute bolus(43,44). A transient increase in the peak inspiratory pressure is believed to overcome acute airway obstruction and minimize cardiorespiratory disturbances. Although surfactant has been given slowly via a syringe pump over 30 minutes or longer(45), this is no longer recommended as a slow infusion has been shown to result in very ununiformed surfactant. distribution and poorer response than bolus treament(46).

Ultrasonic nebulized surfactant treatment has been studied in preterm lambs(47). Compared with instilled surfactant, nebulized surfactant has a lower efficiency of deposition in the non-compliant lung, and a less homogeneous distribution. The potential for nebulized surfactant therapy may therefore be limited. Surfactant has been administered intra-amniotically to four human fetuses in utero with immature amniotic fluid indices(48). After birth, all the infants had an uneventful clinical course without respiratory distress. This was the only report on the possibility of using surfactant antenatally in the prevention of RDS.

Prophylactic versus rescue therapy

Surfactant can be given as prophylactic therapy (at or within 30 minutes of birth to those infants at risk of developing RDS, if possible even before the infant has breathed or received positive pressure ventilation) or as rescue therapy (given only when the diagnosis of severe RDS could be accurately made, usually at 3-6 hours after birth). The advantages of prophylactic over rescue therapy are that it may facilitate initial lung aeration and re- sorption of lung liquid, improve distribution of surfactant administered, and reduce barotrauma and thus leakage of inhibitor proteins. Eight RCTs involving over 2000 infants have -been published on"prophylac- tic versus rescue therapy(49-56). Three meta-analyses of 4 to 7 of these trials revealed a lower incidence of PTX and mortality with prophylactic therapy(57-59). These findings are relevant for infants born extremely preterm, as the majority of these RCTs exclusively 'enrolled infants < 30 weeks gestation. One of these RCTs also reported a reduced risk of severe IVH and ROP(55). However, the risk of chronic lung disease (CLD), or either CLD or death, does not differ according to treatment strategy. No neurodevelopmental advantage has been found on long-term follow-up with prophylactic therapy(60). The disadvantages of prophylactic therapy are that it destabilises the infant during initial resuscitation and that it may be an unnecessary treatment in some infants (thus leading to increased cost, increased risk of side effects and unnecessary endotracheal intubation). A prophylaxis policy would require nearly twice the number of infants treated(58) but result in a 39% reduction in neonatal mortality and an estimated saving of seven more infants for every 100 born < 32 weeks(59). Although there are advantages to prophylactic therapy among those born < 30-32 weeks gestation, the practical disadvantages result in a reluctance to routinely intubate and administer surfactant to all infants born of a certain gestation or birthweight.

It has been recommended that all infants < 32 weeks gestation should be treated with surfactant as soon as they are intubated(59). This would mean surfactant given at birth for such infant who need endotracheal intubation for resuscitation at delivery, or given as Soon as such infants require intubation for ventilation subsequently. Prophylactic therapy at birth has also been advocated for the most immature infants < 28 weeks or < 1000 g who had received no antenatal corticosteroid therapy(53). Delivery room administration of surfactant should be carried out only by someone experienced in neonatal resuscitation and surfactant administration. Other- wise, surfactant therapy should be given as soon as feasible when clinical signs of RDS develop, using the need for endotracheal intubation for mechanical ventilation as the criterion for surfactant administration(61). A protocol of early treatment (less than 2 hours of age in infants at high risk of RDS) compared to late treatment (at about 3 hours when RDS has established) has been shown in a RCT to result in significant 6% reduction in the risk of death or CLD(62). Delaying treatment until the infant has established RDS reduces the efficacy of surfactant therapy. The study showed that early therapy to an estimated 32 infants, when compared with therapy of established RDS, would prevent one infant from death and one infant from developing CLD, but would entail the additional use of surfactant in 8 infants.

In infants with moderate to severe RDS treated early with nasal continuous positive airway pressure (CP AP), administration of a faster acting natural surfactant during a short endotracheal intubation has been shown to reduce the need for subsequent mechanical ventilation (43% vs 85%)(62). Even when a synthetic surfactant was used with CPAP, it has been found to result in a reduction in the. duration of oxygen therapy and days of hospitalization(63). These studies demonstrated the benefits of surfactant therapy in infants treated with nasal CP AP rather than intubated for mechanical ventilation.

High Versus Low Dose

Preterm infants with RDS have a surfactant pool of 5 mg/kg. The surfactant dose is based empirically on animal experiments(64). In term newborn animals, the surfactant pool is about 100 mg/kg which is the usual dose used in surfactant therapy (100mg/kg in 3-5 ml/kg saline). One study using Surfacten from Japan showed that a single dose of 120 mg/kg, compared to 60 mg/kg, resulted in a shorter duration of oxygen and ventilatory therapy and a lower incidence of IVH and CLD(65). An- other study using another bovine surfactant, Alveofact, showed that 100 mg/kg is better than 50mg/kg, with significant improved oxygenation and a lower incidence of pulmonary interstitial emphysema(66). The instillation of surfactant down the en- dotracheal tube is generally well tolerated, especially with the first dose soon after birth and the infant is very sick and requires high peak inspiratory pressures which drives the surfactant rapidly into the lungs. Paradoxically, subsequent doses are less well tolerated because of airway obstruction, since the infants would have been stabilized on lower peak inspiratory pressures by then.

Single Versus Multiple Doses

Only about 20-30% of the surfactant dose can be recovered from the air spaces after 24 hours of ventilation(67). Most of it is found in the lung tissue, being recycled to maintain endogenous metabolic pathways with a turnover time of about 13 hours. Exogenous surfactant does not inhibit the synthesis and secretion of endogenous surfactant. It is of benefit to the surfactant deficient lung by providing it with phospholipids as substrate for the recycling pathways. The improvement after a single dose of surfactant is unsustained because its function can be inhibited by proteins in the small airways and al- veoli(68,69). Multiple doses can be used to overcome this functional inactivation. Three RCTs have shown that multiple doses, compared to a single dose of surfactant, resulted in a decrease in oxygen and ventilatory requirements, PTX and mortality(70-73). Clinical experience suggested that infants treated with a third dose were more preterm and had more neonatal complications such as patent ductus arteriosus (PDA)(73). A RCT has shown that a treatment regime which permits up to four doses has no additional benefit compared to a two-dose regime(61). Although a multiple dose strategy has been shown to be more effective, the optimal timing and indications of re-treatment remain uncertain.

Synthetic Versus Natural Surfactant

The difference between natural and synthetic surfactants appears to be that the synthetic surfactants have a delayed and modest effect 12-18 hours after administration, while oxygenation and lung function improves sooner and to a greater extent after naturalsurfactants(74). Lung function studies showed that the improvement in oxygenation after surfactant instillation is associated with an increase in FRC(74-76). However, even with the use of natural surfactants, dynamic lung compliance does not improve immediately(77-80) but early improvement in static lung compliance was found to correlate with increased oxygenation and recruitment of lung volume(75,81). In contrast, improvement in lung compliance and FRC occurs much later with the use of synthetic surfactants, usually following the clinical improvement in gas exchange(82-84), although pulmonary artery pressure falls by an hour after administration(44,85). It is now generally accepted that surfactant therapy improves aeration of the lung by stabilizing gas exchange units already being ventilated in addition to recruiting new units(74,86). There is probably an improvement in the efficiency in gas mixing and an increase in effective pulmonary blood flow to account for the improvement in oxygenation(74,86). Meta-analyses of surfactant RCTs(5,6) have shown that both synthetic and natural surfactants, whether given as prophylactic or rescue therapy, are effective in reducing the PTX and mortality: rate. Rescue therapy using synthetic surfactant and prophylactic therapy using natural surfactant appeared also to be effective in decreasing the incidence of CLD. Meta-analysis also suggested that the incidence of PDA with synthetic surfactant increased with prophylactic therapy and decreased with rescue therapy but it was not affected by natural surfactant. Although the problem of PDA with surfactant therapy remains controversial, it has been shown that surfactant reduces pulmonary vascular resistance over hour after administration, resulting in a decrease in pulmonary artery pressure(85) and an increase in the velocity of the left-to-right shunting through the PDA(87). In the presence of a large PDA after surfactant therapy, it has been shown that the heart of the preterm infant can mount a compensatory increase of cardiac output sufficient to maintain cerebral blood flow, but postductal organ blood flow cannot be maintained(88). A large left-to-right ductal shunt may lead to hemorrhagic pulmonary edema or "pulmonary hemorrhage". A retrospective study showed that pulmonary hemorrhage was associated with the presence of a clinical detectable PDA be- fore or at the time of the hemorrhage(89). Clinical pulmonary hemorrhage has been reported to increase significantly from 1% to 2% with surfactant therapy though the incidence of pulmonary hemorrhage at necropsy was reported to be similar in treated and control infants(90). Prophylactic use of prostaglandin inhibitors after surfactant therapy within 4 hours after birth has been shown to reduce the incidence of a hemodynamically significant PDA(91).
Nine RCTs involving over 3000 infants, of which three are large multicentre trials(92-94), compared the efficacy of a synthetic surfactant (Exosurf) and natural surfactant (Survanta and Infasurf). Meta-analysis of these studies(58,95) demonstrated with the use of natural surfactant, the oxygen requirement and mean airway pressure are lower for at least.3 days after treatment, the risks of PTX, ROP, neonatal mortality and the combined endpoint of death or CLD are lessened, and fewer doses are required. The use of natural surfactant instead of synthetic one was estimated to prevent one PTX for every 14 infants treated and to prevent one death for every 42 infants treated(96}.

There is some evidence that natural surfactants differ in their effects and are not equivalent. RCTs have shown that Curosurf and Infasurf, compared with Survanta, resulted in more rapid improvement in oxygenation and mean airway pressures up to 48 hours after treatment and a longer duration of effect, with a non-significant trend towards less PTX and large IVH(97,87). No significant long term benefits have yet been established. It is possible that the different surfactant preparations have different physical properties and physiological effects under in vitro and in vivo conditions(99). In the Curosurf study, its beneficial effects over Survanta might also have been explained by the fact the different initial dose of phospholipids used, namely 200 mg/kg for Curosurf and 100 mg/kg for Survanta.

Reasons for Non-Responsiveness

Up to 20% of infants believed to have RDS have little or no response to surfactant therapy. The reasons are multifactorial: (a) extremely preterm infants also have structural lung immaturity, (b) some may have other diseases such as pneumonia or pulmonary hypoplasia, (c) perinatal asphyxia is associated with a poor response, (d) pulmonary edema from lung damage or fluid overload results in protein inhibition and inactivation of surfactant, (e) pulmonary edema from left-to-right shunting through the PDA has the same effect, and if) there may be maldistribution of surfactant in the lungs, though this is not believed to be a common cause of a poor response(100). Clinical studies have shown that a poor response to surfactant is associated with severe RDS, perinatal asphyxia, infection, early PDA, pulmonary interstitial emphysema, PTX and excessive fluid and colloid intake(100-104). Extreme prematurity is not a factor for poor response as surfactant is known to, improve survival without increasing the proportion of disabled survivors in infants < 750 g born at. 23-24 weeks(105,106). It has been shown that infants with a poor immediate response to surfactant therapy has a higher mortality and PTX rates than the good responders(107-108).

Cost Implication

Surfactant is expensive. Nevertheless, surfactant therapy has been shown to result in a 25% reduction in daily hospital charges, a 52% reduction in daily ancillary charges (laboratory, x-ray, respiratory therapy) and, as a result of improved survival, a 22% reduction in hospital charges per survivor(109). It was reported that the cost of care with surfactant therapy declined by 10% among infants who survived and 30% among those who died(8). the lower cost of care. found among those who died shows that the availability of surfactant did not prolong dying for those in whom neonatal intensive care was futile. However, the improved survival rate also increases the average hospital stay, resulting in a cost per extra survivor of 14,000 pounds Sterling(110). Nevertheless, the latter study showed that when the cost per quality adjusted life year was calculated for surfactant therapy, it is one-half that for renal transplantation and one-tenth for that of coronary bypass surgery and hemodialysis.

The cost-effectiveness of surfactant therapy depends on the price of surfactant and whether it is used in large or small preterm infants(111). In the larger infants, surfactant therapy reduces medical costs because it shortens the duration of assisted ventilation and decreases the complications of RDS. In the smaller infants, their increased survival rate prolongs the need for neonatal intensive care among the additional survivors and increases overall medical costs. The savings from the use of surfactant in the larger infants should more than pay for the higher costs incurred in using surfactant in the smaller infants. Therefore, surfactant saves lives, reduces morbidity, and overall also saves money.

Combination Therapy

Non-randomized studies of combined antenatal corticosteroid therapy and surfactant therapy versus surfactant therapy alone have shown that combined therapy resulted in a significant reduction in the incidence of RDS, airleak, PDA, IVH and mortality(112-115). Although postnatal surfactant therapy by itself does not reduce the risk of IVH, antenatal corticosteriod therapy has been shown to have a protective effect on the brain unrelated to enhanced lung maturation(116). Data from animal experiments also indicate that antenatal corticosteroids and postnatal surfactant therapy have synergistic beneficial effects on neonatal lung function(117). Such combined therapy is estimated to result in 125 extra survivors out of 1000 infants born below 30 weeks gestation(118). The study also showed that the cost implications of combined therapy are that 7-11% extra intensive care beds are required. Further- more, 12-24% more special care beds are required after each type of intervention. Nevertheless, the combination of antenatal corticosteroid therapy and postnatal surfactant therapy is the most cost-effective because it produces the greatest number of survivors and the lowest number of intensive and special care days in hospital.

One study in preterm lambs has shown that surfactant therapy and subsequent high-frequency oscillatory ventilation (HFOV) leads to better oxygenation and alveolar expansion at comparable mean airway pressures compared to surfactant therapy followed by conventional mechanical ventilation(119). Another study in preterm monkeys found that the use of surfactant with HFOV, compared to either surfactant or HFOV alone, reduced alveolar proteinaceous edema(120). This reduction in lung injury may be beneficial in lowering the incidence of severity of CLD. The findings of the two studies await confirmation by RCTs in human preterm neonates.

Surfactant for Other Indications

Surfactant deficiency may be present in term infants with respiratory failure due to meconium aspiration syndrome (MAS), pneumonia and the adult type of acute RDS. Meconium inhibits the surface lowering properties of surfactant(121) and surfactant improves lung function in animals with experimentally induced MAS(122). When surfactant was 'given to a series of infants with MAS, it was reported that their oxygenation improved significantly within 2 hours, although 40% showed little or no response(123). The only RCT reported that surfactant-treated infants had improved oxygenation, less time on ventilation, fewer PTX, and less likely to reach the criteria for extracorporeal membrane oxygen- ation(124). Infants with MAS have been treated with tracheal bronchial lavage, using 15 ml/kg of natural surfactant saline suspension at a phospholipid concentration of 5 mg/ml(125). Large RCTs have been proposed to determine whether such a treatment has a role in the management of MAS. Several infants with respiratory failure from bacterial sepsis have also been treated with surfactant which resulted in a reduction in oxygen requirement and mean airway pressure, without apparent side effects(126).

 

 References


1. Fujiwara T, Maeta H, Chida S, Morta T, Watabe Y, Abe T. Artificial surfactant therapy in hyaline membrane disease. Lancet 1980; 1: 55-59.

2. Hall SB. Venkitaraman AR, Whitsett JA, Holm B, Notter RH. Importance of hydrophobic apoproteins as constituents of clinical exogenous surfactants. Am Rev Respir Dis 1992; 145: 24-30.

3. Johansson J, Curstedt T, Robertson B. Synthetic protein analogues in artificial surfactants. Acta Pediatr 1996; 85: 642- 646.

4. Cochrane CG, Revak SD, Merritt T A, Heldt GP, Hallman M, Cunningham MD, et al. The efficacy and safety of KL4 surfactant in preterm infants with respiratory distress syndrome. Am J Respir Crit Care Med 1996; 153: 404-410.

5. SolI RF, McQueen MC Respiratory distress syndrome. In: Effective Care of the Newborn Infant. Eds. Sinclair JC, Bracken MB. Oxford: Oxford University Press, 1992; pp 325-358.

6. Jobe AH. Pulmonary surfactant therapy. New Engl J Med 1993; 328: 861-868.

7. Horbar JD, Wright EC, Onstad L, and members of the National Institute of Child Health and Human Development Neonatal Research Network. Decreasing mortality associated with the introduction of surfactant therapy: An observational study of neonates weighing 601-1300 grams at birth. Pediatrics 1993; 92: 191- 196.

8. Schwartz RM, Luby AM, Scanlon JW, Kellogg RJ. Effect of surfactant on morbidity, mortality, and resource use in newborn infants weighing 500 to 1500g. N'Engl J Med 1994; 330: 1476-1480.

9. McCord FB, Curstedt T, Halliday HL, McClure G, Reid MM, Robertson B. Surfactant treatment and incidence of intra-ventricular hemorrhage in severe respiratory distress syndrome. Arch Dis Child 1988; 63: 10-16.

10. Fujiwara T, Konishi M, Chida S, Watabe Y, Abe T. Surfactant replacement therapy with a single postventilatory dose of a re-constituted bovine surfactant in preterm neonates with respiratory distress syndrome: Final analysis of a multicenter, double-blind, randomized that and comparison with similar trials. Pediatrics 1990;86:753-764.

11. Horbar JD, SolI RF, Schachinger H, Kewitz G, Versmold HT, Lindner W, et al. AEuropean multicentre randomized controlled trial of single dose surfactant therapy for idiopathic respiratory distress syndrome. Eur J Pediatr 1990; 149: 416- 423.

12. Hellstrom-Westas L, Bell AH, Skov L, Greisen G, Svennigson NW. Cerebroelectrical depression following surfactant treatment in preterm neonates. Pediatrics 1992; 89: 643-647.

13. Lundstrom KE, Greison G. Changes in EEG, systemic circulation and blood gas parameters following two or six aliquots of porcine surfactant. Acta Paediatr 1996; 85: 708-712.

14. Cowan F, Whitelaw A, Wertheim D, Silverman M. Cerebral blood flow velocity changes after rapid administration of surfactant. Arch Dis Child 1991; 66: 1105- 1109.

15. Gunkel JH, Banks PLC. Surfactant therapy and intracranial hemorrhage: Review of the literature and results of new analyses. Pediatrics 1993; 92: 775-786.

16. Edwards AD, McCormick DC, Roth SC, Elwell CE, Peebles DM, Cope M, et al. Cerebral hemodynamic effects of treatment with modified natural surfactant investigated by near infrared spectorscopy. Pediatr Res 1992; 32: 532-536.

17. Tubman TR, Rankin SJ, Halliday HL, Johnston S5. Surfactant replacement therapy and the prevalence of acute retinopathy of prematurity. Bioi Neonate 1992; 61(Suppll:): 54-58.

18. Repka MX, Hardy RJ, Phelps DL, Summers CG. Surfactant prophylaxis and retinopathy of prematurity. Arch Ophthalmol1993; 111: 618-620.

19. Termote J, Schalij-Delfos NE, Cats BP, Wittebol-Post D, Hoogervorst BR, Browers HAA. Less severe retinopathy of prematurity induced by surfactant re- placement therapy. Acta Paediatr 1996; 85: 1491-1496.

20. Halliday HL, Mcclure G, Reid McC. Growth and development two years after artificial surfactant replacement at birth. Early Hum Dev 1986; 113: 323-327.

21. Vaucher YE, Merritt TA, Hallman M, Jarvenpaa A, Telsey AM, Jones BL. Nuerodevelopmental and respiratory outcome in early childhood after human surfactant treatment. Am J Dis Child 1988; 142: 927-930.

22. Dunn MS, Shennan AT, Hoskins EM, Lennox K, Enhorning G. Two-year follow-up of infants enrolled in a randomized trial of surfactant replacement therapy for prevention of neonatal respiratory distress syndrome. Pediatrics 1988; 82: 543-547.

23. Morley CJ, Morley R. Follow up of premature babies treated with artificial surfactant (ALEC). Arch Dis Child 1990; 65: 667-669.

24. Ware J, Taeusch HR, SolI RF, McCormick MC Health and developmental outcomes of a surfactant controlled trial: Follow-up at 2 years. Pediatrics 1990; 85: 1103-1107.

25. Robertson B, Curstedt T, Tubman R, Strayer D, Berggren P, Kok J, et al. A 2- year follow up of babies enrolled in a European multicentre trial of porcine surfactant replacement for severe neonatal respiratory distress syndrome. Collaborative European Multicentre Study Group. Eur J Pediatr 1992;151: 372-376.

26. Walti H, Boule M, Moriette G, Relier JP. Pulmonary functional outcome at one year of age in infants treated with natural porcine surfactant at birth. BioI Neonate 1992; 61 (Suppl 1): 48-53.

27. Couser RJ, Ferrara TB, Wheller W, McNamara J, Falde B, Johnson K, Hoekstra RE. Pulmonary follow-up 2.5 years after a randomized, controlled multiple dose bovine surfactant study of preterm newborn infants. Pediatr Pulmonol1993; 15: 163-167.

28. Abbasi S, Bhutani VK, Gerdes JS. Long-term pulmonary consequences of respiratory distress syndrome in preterm infants treated with exogenous surfactant. J Pediatr 1993; 122: 446-452.

29. Survanta Multidose Study Group. Two- year follow-up of infants treated for neonatal respiratory distress syndrome with bovine surfactant. J Pediatr 1994; 124: 962- 967.

30. Corbet A, Long W, Schumacher R, Gerdes J, Cotton R: Double-blind developmental evaluation at 1-year corrected age of 597 premature infants with birth weights from 500 to 1350 grams enrolled in three placebo-controlled trials of prophylactic synthetic surfactant. J Pediatr 1995; 126: S5-S12.

31 Walther FJ, Mullett M, Schumacher R., Sundell H, Easa D, Long W. One-year follow-up of 66 premature infants weighing 500 to 699 grams treated with a single dose of synthetic surfactant or air placebo at birth: Results of a double-blind trial. J Pediatr 1995; 126: S13-S19.

32. Sell M, Cotton R, Hirata T, Gurthie R, LeBlanc M, Mammel M, Long W. One-year follow-up of 273 infants with birth weights of 700 to 1100 grams after pro-phylactic treatment of respiratory distress syndrome with synthetic surfactant or air placebo. J Pediatr 1995; 126: S20-S25.

33. Gerdes J, Gerdes M, Beaumont E, Cook L, Dhanireddy R, Kopleman A, et al. Health and neurodevelopmental outcome at 1- year adjusted age in 508 infants weighing 700 to 1100 grams who received prophylaxis with one versus three doses of synthetic surfactants. J Pediatr 1995; 126: S26- S32.

34. Kraybill EN, Bose C, Corbet A, Garcia-Prats J, Asbill D, Edwards K, Long W. Double-blind evaluation of developmental and health status to age 2 years of infants weighing 700 to 1350 grams treated prophylactically at birth with a single dose of synthetic surfactant or air placebo. J Pediatr 1995; 126: S33-542.

35. Courtney SE, Long W, McMillan D, Walter D, Thompson T, Sauve R, et al. Double-blind 1-year follow-up of 1540 infants with respiratory distress syndrome randomized to rescue treatment with two doses of synthetic surfactant or air in four clinical trials. J Pediatr 1995; 126: S43-S52.

36. Casiro 0, Bingham W, MacMurray B, Whitfield M, Saigal S, Vincer M, Long W. One-year follow-up of 89 infants with birth weights of 500 to 749 grams and respiratory distress syndrome randomized to two rescue doses of synthetic surfactant or air placebo. J Pediatr 1995; 126: S53-S60.

37. Saigal S, Robertson C, Sarikaran K, Bingham W, Casiro 0, MacMurray B, et al. One-year outcome in 232 premature infants with birth weights of 750 to 1249 grams and respiratory distress syndrome randomized to rescue treatment with two doses of synthetic surfactant or air placebo. J Pediatr 1995; 126: 561-567.

38. Gong A, Aanday E, Boros S, Bucciarelli R, Burchfielf D, Zucker J, Long W. One-year follow-up evaluation of260 premature infants with respiratory distress syndrome and birth weights of 700 to 1350 grams randomized to two rescue doses of synthetic surfactant or air placebo. J Pediatr 1995; 126: 568-574.

39. Sauve R, Long W, Vincer M, Bard H, Derleth , Stavenson D, et al. Outcome at 1-year adjusted age of 957 infants weighing more than 1250 grams with respiratory distress syndrome randomized to receive synthetic surfactant or air placebo. J Pediatr 1995; 126: 575-580.

40. Zola EM, Gunkel JH, Chan RK, Urn MO, Knox I, Feldman BH, et al. Comparison of three dosing procedures for administration of bovine surfactant to neonates with respiratory distress syndrome. J Pediatr 1993;122:453-459.

41. Merritt TA, Kheiter A, Cochrane CG. Positive end-expiratory pressure during KL4 surfactant instillation enhances intrapulmonary distribution in a simian model of respiratory distress syndrome. Pediatr Res 1995; 38: 211-217.

42. Broadbent R, Fok TF, Dolovich M, Watts 1, Coates G, Bowen B, Kirpalani H. Chest position and pulmonary deposition of surfactant in sufactant depleted rabbits. Arch Dis Child 1995; 72: F84~F89.

43. Saliba E, Nashashibi M, Vaillant M-C, Nasr C, Laugier J. Instillation rate effects of Exosurf on cerebral and cardiovascular hemodynamics in preterm neonates. Arch Dis Child 1994; 71: FI74-FI78.

44. Skinner J. The effects of surfactant on hemodynamics in hyaline membrane disease. Arch Dis Child 1997; 76: F67-F69.

45. Sitler CG, Turnage CS, McFadden BE, Smith EO, Adams JM. Pump administration of exogenous surfactant: Effects on oxygenation, heart rate, and chest wall movement of premature infants. J Perinatol1993; 13: 197-2000.

46. Ueda T, Ikegami M, Rider ED, Jobe AH. Distribution of surfactant and ventilation in surfactant-treated preterrn lambs. J Appl Physiol1994; 76: 45-55.

47. Henry MD, Rebello CM, Ikegami M, Jobe AH, Langenbaack EG, Davis JM. Ultra-sonic nebulized in comparison with instilled surfactant treatment of preterm lambs. Am J Respir Crit Care Med 1996; 154: 366-375.

48. Cosmi EV, La Torre R, Di Iorie R, Anceschi MM. Surfactant administration to the human fetus in utero: A new approach to prevention of neonatal respiratory distress syndrome. J Perinat Med 1996; 24: 191-193.

49. Dunn MS, Shennan AT, Zayack D, Possmayer F. Bovine surfactant replacement therapy in neonates of less than 30 weeks' gestation: A randomized controlled trial of prophylaxis versus treatment. Pediatrics 1991; 87: 377-386.

50. Kendig JW, Notter RH, Cox C, Reubens LJ, Davis JM, Maniscalco WM, et al. A comparison of surfactant as immediate prophylaxis and as rescue therapy in newborns of less than 30 weeks' gestation. New Engl J Med 1991; 324; 865-871.

51. Merritt T A, Hallman M, Berry C, Pohjavuori M, Edwards DK, Jaaskelainen J, et al. Randomized, placebo- controlled trial of human surfactant given at birth versus rescue administration in very low birth weight infants with lung immaturity. J Pediatr 1991; 118: 581-594.

52. Konishi M, Fujiwara T, Chida 5, Maeta H, Shimada S, Kasai T, et al. A prospective, randomized trial of early versus late ad- ministration of a single dose of surfac- tant-RA. Early Hum Dev 1992; 29: 275- 282.

53. Egberts J, de Winter JP, Sedin G, de Kleine MJK, Broberger U, can Bel F, et al. Comparison of prophylaxis and rescue.

54. treatment with Curosurf in neonates less than 30 weeks' gestation: a randomized.. trial. Pediatrics 1993; 92: 768-774. Kattwinkel J, Bloom BT, Delmore P, Davis CL, Farrell E, Friss H, et al. Prophylactic administration of calf lung surfactant extract is more effective than early treatment of respiratory distress syndrome in neonates of 29 through 32 weeks' gesta- tion. Pediatrics 1993; 92: 90-98.

55. Walti H, Paris-Llado J, Breart G, Couchard M, and the French Collabora- tive Multicentre Study Group. Porcine surfactant replacement therapy in newborns of 25-31 weeks' gestation: A randomized, multicentre trial of prophylaxis versus rescue with multiple low doses. Acta Paediatr 1995; 84: 913-921.

56. Bevilacqua G, Parmigiani S, Robertson B and the Italian Collaborative Multicenter Group. Prophylaxis of respiratory diestress syndrome by treatment with modified porcine surfactant at birth: A multicenter prospective randomized trial. J Perinat Med 1996; 24: 609-620.

57. Dunn MS. Surfactant replacement therapy: Prophylaxis or treatment? Pediatrics 1993; 92: 148-150.

58. Roll RF. Surfactant therapy in the USA: Trials and current routines. BioI Neonate 1997;71: 1-7.

59. Morley CJ. Systematic review of prophylactic vs rescue surfactant. Arch Dis Child 1997; 77: F70-F74.

60. Vaucher YE, Harker L, Merritt TA, Hallman M, Gist K, Bejar R, et al. Outcome at twelve months of adjusted age in very low birth weight infants with lung immaturity: A randomized, placebo-con- trolled trial of human surfactant. J Pediatr 1993; 122: 126-132.

61. The OSIRIS Collaborative Group. Early versus delayed neonatal administration of synthetic surfactant-the judgement of OSIRIS. Lancet 1992; 340: 1363-1369.

62. Verder H, Robertson B, Greisen G, Ebbesen T, Albertsen P, Lundstrom K, et al. Surfactant therapy and nasal continuous positive airway pressure for newborns with respiratory distress syndrome. N Engl J Med 1994; 331: 1051-1055.

63. Alba J, Agrawal R, Hegyi T, Hiatt 1M. Efficacy of surfactant therapy in infants managed with CP AP. Pediatr Pulmonol 1995; 20: 172-176.

64. Ikegami M, Adams FH, Towers B, Osher AB. The quantity of natural surfactant necessary to prevent the respiratory distress syndrome in premature lambs. Pediatr Res 1980; 14: 1082-1085.

65. Konishi M, Fujiwara T, Naito T, Takeuchi Y, Ogawa Y, Inukai K, et al. Surfactant replacement therapy if). neonatal respiratory distress syndrome. A multicentre, randomized clinical trial: Comparison of high-versus low-dose surfactant T A. Eur J Pediatr 1988; 147: 20-25.

66. Gortner L, Pohlandt F, Bartmann P. High-dose versus low-dose bovine surfactant treatment in very prematur infants. Acta Paediatr 1994; 83: 135-141.

67. Jobe AH, Ikegami M, Seidner SR, Pettenazzo A, Ruffini L. Surfactant phosphotidylcholine metabolism and surfactant function in preterm ventilated lambs. Am Rev Respir Dis 1989; 139: 352- 359.

68. Fuchimukai T, Fujiwara T, Takahashi A, Enhorning G. Artificial pulmonary surfactant inhibited by proteins. J Appl Physiol1987; 62: 429-437.

69. Hallman M, Merritt TA, Akino T, Bry K. Surfactant protein A, phosphatidylcholine, and surfactant inhibitors in epithelial lining fluid: Correlation with surface activity, severity of respiratory distress syndrome, and outcome in small premature infants. Am Rev Respir Dis 1991; 144: 1376-1384.

70. Dunn MS, Shennan AT, Possmayer F. Single-versus multiple-dose surfactant replacement therapy in neonates of 30 to 36 weeks' gestation with respiratory distress syndrome. Pediatrics. 1990; 86: 564-571.

71. Speer CP, Robertson B, Curstedt T, Halliday HL, Compagnone D, Gefeller , et al. Randomized European multicenter trial of surfactant replacement therapy for severe neonatal respiratory distress syndrome: Single versus multiple doses of Curosurf. Pediatrics 1992; 89: 13-20.

72. Corbet A, Gerdes J, Long W, Avila E, Puri A, Rosenberg A, et a/. Double-blind, randomized trial of one versus three prophylactic doses of synthetic surfactant in 826 neonates weighing 700 to 1100 grams: Effects on mortality rate. J Pediatr 1995; 126: 969-978.

73. Easa D, Pelke S, Nakamura KT, Barrett J, Balaraman V, Loo SWH, et al. Exosurf treatment investigational new drug phase: Effect of an individualized third dose in infants with respiratory distress syndrome. Pediatr Pulmonol1992; 14: 16- 22.

74. Cotton RB, Olsson T, Law AB, Parker RA, Lindstrom DP, Siiberberg AR, et al. The physiologic effects of surfactant treatment on gas exchange in newborn premature infants with hyaline membrane disease. Pediatr Res 1993; 34: 495-501.

75. Goldsmith LS, Greenspan JS, Rubenstein SD, Wolfson MR, Shaffer, TH. Immediate improvement in lung volume after exogenous surfactant: alveolar recruitment versus increased distention. J Pediatr 1991; 119: 424-428.

76. Svenningsen NW, Bjorklund L, Vilstrup C, Werner O. Lung mechanics (FRC and static pressure-volume diagram) after en- dotrachealsurfactant instillation: preliminary observations. BioI Neonate 1992; 61 (Suppl 1): 44-47.

77. Davis JM, Veness-Meehan K, Notter RH, Bhutani VK, Kendig JW, Shapiro. Changes in pulmonary mechanics after the administration of surfactant to infants with respiratory distress syndrome. New Engl J Med 1988; 319: 476-479.

78. Couser RJ, Ferrara TB, Ebert J, Hoekstra RE, Fangman JJ. Effects of exogenous surfactant therapy ondynamic compliance during mechanical breathing in preterm infants with hyaline membrane disease. J Pediatr 1990; 116: 119-124.

79. Kelly E, Bryan H, Possmayer F, Frndova H, Bryan C. Compliance of the respiratory system in newborn infants pre-and postsurfactant replacement, therapy. Pediatr Pulmonol1993; 15: 225-230.

80. de Winter JP, Merth IT, van Bel F, Egberts J, Brand R, Quanjer PhH. Changes of respiratory system mechanics in ventilated lungs of preterm infants with two different schedules of surfactant treatment. Pediatr Res 1994; 35: 541-549.

81. Baraldi E, Pettenazzo A, Filippone M, Magagnin GP, Saia OS, Zacchello. Rapid improvement of static compliance after surfactant treatment in preterm infants with respiratory distress syndrome. Pediatr Pulmonol1993; 15: 157-162.

82. Pfenninger J, Aebi C, Bachmann D, Wagner BP. Lung mechanics and gas exchange in ventilated preterm infants during treatment of hyaline membrane disease with multiple doses of artificial surfactant (Exosurf). Pediatr Pulmonol 1992; 14: 10-15.

83. Armsby DH, Bellon G, Carlisle K, Rector D, Baldwin R, Long W, et a/. Delayed compliance increase in infants with respiratory distress syndrome following synthetic surfactant. Pediatr Pulmonol 1992; 14: 206-213.

84. Corcoran JD, Berggren P, Sun B, Halliday HL, Robertson B, Curstedt T. Comparison of surface properties and physiological effects of a synthetic and a natural surfactant in preterm rabbits. Arch Dis Child 1994; 71: F165-169.

85. Hamdan AH, Shaw NJ. Changes in pulmonary artery' pressure in infants with respiratory distress syndrome following treatment with Exosurf. Arch Dis Child 1995;72:F176-179.

86. Milner AD. How does exogeneous surfactant work? Arch Dis Child 1993; 68: 253- 254.

87. Kaapa P, Seppanen M, Kero P, Saraste M. Pulmonary hemodynamics after synthetic surfactant replacement in neonatal respiratory distress syndrome. J Pediatr 1993; 123: 115-119.

88. Shimada S, Kasai T, Konishi M, Fujiwara T. Effects of patent ductus arteriosus on left ventricular output and organ blood flows in preterm infants with respiratory distress syndrome treated with surfactant. J Pediatr 1994; 125: 270-277.

89. Garland J, Buck R, Weinberg M. Pulmonary hemorrhage risk in infants with a clinically diagnosed patent ductus arteriosus: A retrospective cohort study. Pediatrics 1994; 94: 719-723.

90. van Houten J, Long W, Mullet M, Finer N, Derieth D, McMurray B, et al. Pulmonary hemorrhage in premature infants after treatment with synthetic surfactant: An autopsy evaluation. J Pediatr 1992; 120: 540-544.

91. Amato M, Huppi P, Markus D. Prevention of symptomatic patent ductus arteriosus with ethamsylate in babies treated with exogenous surfactant. J Perinatol 1993; 13: 2-7.

92. Horbar JD, Wright LL, SolI RF, Wright EC, Fanaroff AA, Korones SB, et al. A multicenter randomized trial comparing two surfactants for the treatment of neonatal respiratory distress syndrome. J Pediatr 1993; 123: 757-766.

93. Vermont-Oxford Neonatal Network. A multicentet, randomized trial comparing synthetic surfactant with modified bovine surfactant extract in the treatment of neo- natal respiratory distress syndrome. Pediatrics 1996; 97: 1-6.

94. Hudak M, Farrell EE, Rosenberg AA, Jung AL. Auten RL, Durand DJ, et al. A multicenter randomized, masked comparison trial of natural versus synthetic surfactant for the treatment of respiratory distress syndrome. J Pediatr 1996; 128: 396-406.

95. Halliday HL. Synthetic or natural surfactants. Acta Paediatr 1997; 86: 233-237.

96. Halliday HL. Clinical trials of surfactant replacement in Europe. BioI Neonate 1997; 71 (Suppll): 8-12.

97. Speer CP, Gefeller O, Groneck P. Randomized clinical trial of two treatment regimens of natural surfactant preparations in neonatal distress syndrome. Arch Dis Child 1995; 72: F8-F13.

98. Bloom BT, Kattwinkel J, Hall RT, Delmore PM, Egan EA, Trout JR, et al. Comparison of Infasurf (calf lung surfactant extract) to Survanta (Beractant) in the treatment and prevention of respiratory distress syndrome. Pediatrics 1997; 100: 31-38.

99. Cummings JJ, Holm BA, Hudak ML, Hudak ML, Hudak BB, Ferguson WH, Egan EA. A controlled clinical comparison of four different surfactant preparations in surfactant-deficient preterm lambs. Am Rev Respir Dis 1992; 145: 999-1004.

100. Charon A, Taeusch HW, Fitzgibbon C, Smith GB, Treves ST, Phelps DS. Factors associated with surfactant treatment response in infants with severe respiratory distress syndrome. Pediatrics 1989; 83: 348-354.

101. Collaborative European Multicentre Study Group. Factors influencing the clinical response to surfactant replacement therapy in babies with severe respiratory distress syndrome. Eur J Pediatr 1991;150:433-439.

102. Konishi M, Chida S, Simada S, Kasai T, Murakami Y, Cho K, et al. Surfactant replacement therapy in premature babies with respiratory distress syndrome: Factors affecting the response to surfactant and comparison of outcome from 1982-86 and 1987-91. Acta Paediatr Jpn 1992; 34: 617-630.

103. Hallman M, Merritt T A, Bry K, Berry C. Association between neonatal practices and efficacy of exogenous human surfactant: Results of bicenter randomized trial. Pediatrics 1993; 91: 552-560.

104. Hamvas A, Devine T, Cole FS. Surfactant therapy identifies infants at risk for pulmonary mortality. Am J Dis Child 1993; 147: 665-668.

105. Ferrara TB, Hoekstra RE, Couser RJ, Jack- son JC, Anderson CL, Myers TF, Raye JR. Effects of surfactant therapy on outcome of infants with birth weights of 600 to 750 grams. J Pediatr 1991; 119: 455-457.

106. Ferrara TB, Hoekstra RE, Couser RJ, Gaziano EP, Calvin SE, Payne NR, Fangman JJ. Survival and follow-up of in- fants born at 23 to 26 weeks of gestational age: effects of surfactant therapy. J Pediatr' 1994;124: 119-124.

107. Segerer H, Stevens P, Schadow B, Maier R, Kattner E, Schwarz H, et al. Surfactant substitution in ventilated very low birth weight infants: Factors related to response types. Pediter Res 1991; 30: 591- 596.

108. Kuint J, Reichman B, Neumann L, Shinwell ES. Prognostic value of the immediate response to surfactant. Arch Dis Child 1994; 71: F170-F173.

109. Maniscalco WM, Kendig JW, Shapiro DL. Surfactant replacement therapy: impact on hospital charges for premature infants with respiratory distress syndrome. Pediatrics 1989; 83: 1-6.

110. Tubman TRJ, Halliday HL, Normand C. Cost of surfactant replacement treatment for severe neonatal respiratory distress syndrome: A randomized controlled trial. Brit Med J 1990; 301: 842-845.

111. Mugford M, Piercy J, Chalmers 1. Cost implications of different approaches to the prevention of respiratory distress syndrome. Arch Dis Child 1991; 66: 757-764.

112. Farrell EE, Silver RK, Kimberlin LV, Wolf ES, Dusik JM. Impact of antenatal dexamethasone administration on respiratory distress syndrome in surfactant- treated infants. Am J Obstet Gynecol 1989; 161: 628-633.

113. Jobe AH, Mitchell BR, Gunkel JH. Beneficial effects of the combined use of prenatal corticosteroids and postnatal surfactant on preterm infants. Am J Obstet Gyneco11993; 168: 508-513.

114. White A, Marcucci G, Andrews E, Edwards K, Long W. Antenatal steroids and neonatal outcomes in controlled clinical trials of surfactant replacement. Am J Obstet Gyneco11995; 173: 286-290.

115. Andrews EB, Marcucci G, White A, Long W. Associations between use of antenatal corticostewids and neonatal outcomes within the Exosurf neonatal treatment in- vestigational new drug program. Am J Obstet Gynecol1995; 173: 290-295.

116. Leviton A, Kurban KC, Pagano M, Allred EN, Van Marter L. Antenatal corticosteroids appear to reduce the risk of postnatal germinal matrix hemorrhage in intubated low birth weight newborns. Pediatrics 1993;91:1083-1088.

117. Robertson B. Corticosteroids and surfactant for prevention of neonatal RDS. Ann Med 1993; 25: 285-288.

118. Egberts J. Estimated costs of different treatments of the respiratory distress syndrome in a large cohort of preterm infants to less than 30 weeks of gestation. BioI Neonate 1992; 61 (Suppl1): 59-65.

119. Walther FJ, Kuipers 1M, Gidding CEM, Willebrand D, Buchholtz RTF, Bevers EM. A comparison of high-frequency oscillation superimposed onto back up mechanical ventilation and conventional mechanical ventilation on the distribution of exogenous surfactant in premature lambs. Pediatr Res 1987; 22: 725-729.

120. Jackson JC, Truog WE, Standaert TA, Murphy JH, Juul SE, Chi EY, et al. Reduction in lung jury after combined surfactant and high-frequency ventilation. Am J Respir CritCare Med 1994; 150: 534-539.

121. Sun B, Curstedt T, Robertson B. Surfactant inhibition in experimental meconium aspiration syndrome. Acta Paediatr 1993; 82: 182-189.

122. Sun B. Exogenous surfactant improves ventilation efficiency and alveolar expansion in rats with meconium aspiration. Am J Respir Crit Care Med 1996; 154: 764- 770.

123. Halliday HL, Speer CP, Robertson B. Treatment of severe meconium aspiration syndrome with porcine surfactant. Eur J Pediatr 1996; 155: 1047-1051.

124. Findley RD, Taeusch HW, Walther FJ. Surfactant replacement therapy for meconium aspiration syndrome. Pediatrics 1996; 97:48-52.

125. Ibara S, Ikenoue T, Murata Y, Sakamoto H, Saito T, Nakamura Y, et al: Management of severe meconium aspiration syndrome by tracheobronchial lavage and replacement of Surfactant- T A. Acta Paediatr Japon 1995; 37: 64-67.

126. Fetter WPF, Baerts W, Bos AP, van Lingen RA. Surfactant replacement therapy in neonates with respiratory failure due to bacterial sepsis. Acta Paediatr 1995; 84: 14-16.

Home

Past Issue

About IP

About IAP

Feedback

Links

 Author Info.

  Subscription