Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Apr 1.
Published in final edited form as: Semin Perinatol. 2016 Jan 29;40(3):189–202. doi: 10.1053/j.semperi.2015.12.006

PHARMACOLOGIC INTERVENTIONS FOR THE PREVENTION AND TREATMENT OF RETINOPATHY OF PREMATURITY

Kay D Beharry 1,2,3, GB Valencia 1, DR Lazzaro 2,3, JV Aranda 1,2,3
PMCID: PMC4808450  NIHMSID: NIHMS755875  PMID: 26831641

Abstract

Retinopathy of prematurity (ROP), a significant morbidity in prematurely born infants, is the most common cause of visual impairment and blindness in children and persists till adulthood. Strict control of oxygen therapy and prevention of intermittent hypoxia are key in the prevention of ROP, but pharmacologic interventions have decreased risk of ROP. Various drug classes such as methylxanthines (caffeine), VEGF inhibitors, anti-oxidants, and others have decreased ROP occurrence. The timing of pharmacologic intervention remains unsettled, but early prevention rather than controlling disease progression may be preferred. These drugs act through different mechanisms and synergistic approaches should be considered to maximize efficacy and safety.

INTRODUCTION

Retinopathy of prematurity (ROP) is a developmental vascular disorder characterized by abnormal growth of retinal blood vessels in the incompletely vascularized retina of extremely low gestational age neonates (ELGANs) who are <1250 grams, < 28 weeks gestation (13). In the United States, ROP afflicts about 16,000 ELGANs annually (1), and remains the third leading cause of childhood blindness (14%) with much higher rates in developing countries (5). Incomplete retinal vascularization due to prematurity and oxygen are key factors in ROP, however, the etiology of this “new” form of ROP is multivariate and complex, and involves hypersensitivity of the immature retina to changes in oxygen (4,6,7).

Pathophysiology of ROP

In humans, the retina develops in utero where tissue oxygen is low (7). Vascular precursor cells are laid from 12 to 21 weeks gestational age creating a scaffold for future vessel development. The vessels emerge from the optic disk and follow a VEGF template established by astrocytes which populate the retina before the vessels (8). Angiogenesis begins at approximately 16 to 17 weeks gestational age, with new vessels budding from existing vessels. The metabolic demands of the developing retina exceed the oxygen supplied by the choroidal circulation resulting in ‘physiologic hypoxia,’ and thus stimulate angiogenesis (7). Vasoactive factors, such as insulin-like growth factor (IGF)-1, vascular endothelial growth factor (VEGF) and erythropoietin (Epo), in addition to maternally derived factors, stimulate new vessel formation. The vessels reach the nasal ora serrata by 36 weeks and the temporal ora serrata by 40 weeks. In ELGANs, the retinal vasculature is immature and thus vulnerable to oxidative damage. Early studies by Ashton et al. (9) demonstrated that exposure to oxygen causes vaso-obliteration and vaso-proliferation when room air breathing was resumed. Those early studies led to a two-phase hypothesis of ROP: 1) Phase 1 or vaso-obliteration, begins at preterm birth with the transition from an intrauterine to extrauterine environment causing a rise in PaO2 of 30–35 mm Hg to 55–80 mmHg and loss of placental and maternal growth factors. During this phase, exposure to supplemental oxygen, required for treatment of respiratory distress syndrome, further suppresses retinal growth factors which are already compromised due to preterm birth and poor nutrition (10), thus leading to arrest and retraction of the developing retinal vessels, or vaso-obliteration; and 2) Phase 2 or vaso-proliferation, begins at approximately 32–34 weeks (11). As the infant matures, the avascular retina becomes metabolically active, inducing a second phase, or retinal neovascularization (3). This phase of ROP is driven by hypoxia and subsequent upregulation of VEGF and IGF-I which leads to abnormal vascular overgrowth into the vitreous, retinal hemorrhages, retinal folds, dilated and tortuous posterior retinal blood vessels, or “Plus” disease, and retinal detachment. ELGANs with chronic lung disease experience numerous alterations in their O2 saturations or apneas (12,13). Infants who experience the greatest fluctuations in their PaO2 seem to be at a higher risk for the development of threshold ROP (6,13). In these infants with “new” ROP, intermittent hypoxia (IH) occurs during supplemental oxygen treatment, or Phase 1, thus worsening the outcomes during Phase 2. Indeed this was demonstrated in a rat model which utilized brief episodes of hypoxia during hyperoxia, simulating apnea of prematurity (1417). The fluctuating oxygen model also shows a higher incidence of intravitreal neovascularization (18) with corresponding high levels of retinal VEGF (19) and vitreous fluid growth factors (14,15). The pattern of IH may also play a role in the development of ROP (13) and OIR (1417). Clustering IH episodes resulted in a more severe form of OIR with increased retinal hemorrhages, vascular tufts, leaky vessels, vascular tortuosity, and vascular overgrowth, compared to dispersed IH episodes. This may be due to differences in exposure time of the retina to hypoxia at a given time point. Clustering episodes of brief hypoxia or grouping of desaturations with minimal time for recovery between episodes causes the retina to remain hypoxic for a longer period of time thus leading to a more exaggerated increase in VEGF resulting in characteristics consistent with “Plus” disease (14). In light of these new findings, the Phase 1/Phase 2 hypothesis of ROP originally proposed in 1954 by Ashton et al. (9), may need to be redefined with respect to “new” ROP and IH.

Oxygen

Oxygen is the most commonly used drug in neonatal care for respiratory support (20). The widespread use of unrestricted oxygen in preterm infants began in the early 1940s in response to observations that inspired oxygen improved the irregular breathing pattern of premature infants (21,22). This led to the first epidemic of ROP, described in 1942 by Terry et al. (23) and then known as retrolental fibroplasia or fibroblastic overgrowth behind the crystalline lens.. In 1951, it was suggested that oxygen use was associated with ROP (24). This was confirmed in 1952 in humans (25) and later in animals (26). By 1953, approximately 10,000 infants worldwide were blinded (21). The first multicenter randomized clinical trial to study ROP started in 1953 and involved 18 centers. The study enrolled infants <1500 grams in two arms: 1) FiO2 >50% for 28 days; and 2) FiO2 < 50%. In 1954, one of the centers reported that blindness was prevented if oxygen did not exceed 40%. Six years later, review of autopsies revealed that curtailed use of oxygen increased the incidence of mortality such that for every eye sight gained, 16 lives were lost (Bolton, Lancet 1974). Despite the introduction of transcutaneous oxygen monitoring and pulse oximetry in the 1960s to 1980s, and many nonrandomized and randomized clinical trials, the optimum range of oxygenation in preterm infants remains elusive and controversial. The Phase 1 (hyperoxia)/Phase 2 (hypoxia) hypothesis of ROP led to the premise that administration of oxygen during Phase 2 would increase tissue oxygen, decrease VEGF and curtail vessel overgrowth. This hypothesis was tested in the Supplemental Therapeutic Oxygen for Prethreshold Retinopathy of Prematurity (STOP-ROP) randomized clinical trial in 2000 (28). The study randomized 649 infants with prethreshold ROP and O2 saturation <94% at 35 weeks to oxygen saturation targets of 89–95% or 96–99%. The STOP-ROP study failed to decrease progression of the disease or reduce the number of infants requiring peripheral ablative surgery and showed no differences in the development of threshold ROP. ELGANs experience continuous fluctuations in arterial O2 saturation, therefore increasing inspired oxygen may not be an appropriate approach (29,30). Instead, low and stable oxygen therapy may be more beneficial (2). The Australian Benefits of Oxygen Saturation Targeting (BOOST) randomized, double-blind, multicenter trial involving 358 preterm infants, kept the saturation ranges at 91–94% or 95–98%. The incidence of severe ROP was comparable between the groups (31). Five large multicenter, masked, randomized, control trials (collectively known as The Neonatal Oxygen Prospective Meta-Analysis, or NeOPRoM Collaboration) enrolled approximately 5000 ELGANs <28 weeks, compared 85–89% versus 91–95%. In the United States, the Surfactant Positive Pressure and Pulse Oximetry (SUPPORT) trial reported that SpO2 levels of 85–89% was associated with increased mortality and a higher incidence of severe ROP was found in the 91–95% group (32). This was reflected in the BOOST II trial (Australia, New Zealand and United Kingdom) after interim analysis, and enrollment was stopped (33). The Canadian Oxygen Trial (COT) reported no difference in mortality or severe ROP (34). A recent meta-analysis of all published randomized trials evaluating the effect of restricted versus liberal oxygen exposure in preterm infants show no difference in ROP (35). After over 70 years of oxygen use, and despite large multi-center, randomized clinical trials, there is still no consensus regarding optimal oxygen therapy for ELGANs. Oxygen remains the most commonly used drug in neonatal intensive care units worldwide (36) and the incidence of severe ROP has not appreciably declined.

PHARMACOLOGIC INTERVENTIONS IN ROP

In addition to better control of oxygen therapy, numerous pharmacologic interventions have been tried and some appears promising for the prevention of ROP and stopping its progression. These pharmacologic agents are discussed.

Antioxidants

Oxygen used in excess has damaging effects particularly when used in ELGANs. In normal respiration, oxygen is reduced in the mitochondria to two molecules of water during the process of aerobic energy metabolism, or oxidative phosphorylation (OXPHOS), which produces more than 90% of our cellular energy (37). During OXPHOS, reactive oxygen species (ROS) are produced as a byproduct. ROS are highly reactive chemical molecules that react with lipids to initiate lipid peroxidation and DNA damage (38). Major ROS produced in the mitochondria are superoxide anion, hydrogen peroxide (H2O2), hydroxyl radical, and hydroperoxyl radical. Reactive nitrogen species (RNS) such as nitric oxide (NO), nitrogen dioxide (NO2), dinitrogen trioxide (N2O3), and peroxynitrite, are also produced. The primary endogenous scavengers of ROS, or antioxidants, are superoxide dismutase (SOD), catalase and glutathione peroxidase (Gpx). SOD acts on superoxide anion to produce hydrogen peroxide (H2O2), catalase converts H2O2 to water, and Gpx detoxifies peroxides and hydroperoxides. Other antioxidants include peroxiredoxins (which catabolize H2O2 to water), peroxiredoxin, heme oxygenase, glutaredoxin and thioredoxin, and nonenzymatic agents such as β-carotene, retinol, vitamin C, and vitamin E. When endogenous antioxidant systems are inadequate or overwhelmed by ROS many pathologies including hypoxia/reperfusion injury and ROP develop. The retina is rich in mitochondria and has a high rate of OXPHOS thus rendering it a target for ROS and oxidative damage (39,40). Excessive ROS production contributes to mitochondrial damage and vascular dysfunction, particularly in ELGANs with immature antioxidant systems (4143). Therefore, many investigators tested the hypothesis that antioxidants are beneficial for treatment and/or prevention of ROP.

Vitamin E was the first antioxidant to be used for treatment and/prevention of ROP (44). Several later trials of supplementing preterm infants with vitamin E for preventing or limiting ROP have been conducted with conflicting results (4551). A randomized trial of 755 VLBW infants found no significant decrease of severe ROP although the progression from moderate to severe ROP was less frequent following vitamin E treatment (47). Phelps et al. (52) and Rosenbaum et al. (53) found increased retinal hemorrhages and an increased rate of grade 3 and 4 IVH in the vitamin E group. Two meta-analyses reported beneficial effects of Vitamin E for reducing the risk of ROP and blindness. The earlier report analyzed 6 clinical trials and found a 52% reduction in the incidence of Stage 3+ ROP (54). The later report analyzed 26 randomized clinical trials and a reduction in severe ROP and blindness (55). A third report found no effect at all (56). In this report, the authors analyzed 9 randomized controlled trials and concluded that not more than about 4% of all very low-birthweight infants are likely to benefit from routine vitamin E supplementation. Vitamin E was the most commonly used antioxidant for ROP spanning over 4 decades. Its use has now been largely abandoned secondary to increased risks of morbidity with no conclusive evidence for protection against severe ROP.

Other antioxidants used to treat/prevent ROP include: D-Penicillamine (DPA) an antioxidant and suppressor of VEGF bioavailability (57). DPA was introduced to prevent ROP in the 1980s. In a prospective controlled trial, DPA was administered to preterm infants at 26–35 weeks gestation and found to be effective for ROP with no serious adverse effects (58) and at one year follow-up, (59). Later trials refuted those findings (60,61); Superoxide Dismutase (SOD) dismutates the extremely toxic superoxide anion into H2O2 and H2O. SOD is the first line of defense against oxidative stress in the mitochondria, with MnSOD located in the inner mitochondrial matrix. A multicenter trial of intratracheal rhSOD showed a reduction in severe ROP above stage 2 in rhSOD-treated infants born at <25 weeks (62); Lutein and zeaxanthin are antioxidants in the eye. A single center, double-blind randomized controlled trial comparing lutein administration versus placebo, showed no differences in the incidence of ROP at any stage (63). These findings were later confirmed (64,65); Vitamin A is one of the most important micronutrients necessary for growth and differentiation of tissues (66). Vitamin A supplementation reduces infant mortality (67). Retinal vitamin A allows adequate rhodopsin availability for phototransduction and protects the photoreceptors from the harmful effects of hypoxia and hyperoxia (68). Studies show a trend for decreased incidence of threshold ROP in ELBW infants treated with 10,000 IU intramuscular vitamin A three times a week (0%) compared with 16% in those who received half this dose (69). A recent double-blind, randomized controlled trial of early high-dose intramuscular vitamin A supplementation for infants at risk of ROP showed improves retinal function at 36 weeks' PMA (68). A meta-analysis of the three studies suggests a trend towards reduced incidence of ROP in vitamin A supplemented infants (70). Although promising, there is a paucity of studies examining the benefits of vitamin A for severe ROP. Considering its antioxidant properties and possible beneficial effects on the photoreceptors, further studies are warranted; and Allopurinol is a xanthine oxidase and superoxide anion inhibitor. A randomized, controlled, clinical trial involving 400 preterm infants who received either allopurinol or placebo showed no benefit (71).

Cyclooxygenase Inhibitors

In preterm infants who are exposed to supraphysiologic levels of oxygen, and have compromised antioxidant systems, the imbalance of oxidant/antioxidants in favor of oxidants lead to oxidative stress and activation of proinflammatory mediators, including prostanoids (72, 73). The primary pathway through which prostanoids are activated is through arachidonic acid (AA), a long-chain polyunsaturated fatty acids released from phospholipids through the action of phospholipase A1 (PLA2) (74). AA is the precursor of the eicosanoids, including prostanoids, through the cyclooxygenase (COX) enzyme, and leukotrienes, through the lipoxygenase (LOX) pathway. The two main COX isoforms are the constitutively expressed COX-1, responsible for maintaining basal prostanoid levels, and the inducible COX-2 which is generally absent and is induced in inflammation (75,76). Both isoforms transform AA into prostaglandin H2, which is then transformed by synthases into prostaglandins (PGs) and thromboxanes. A third isoform, COX-3 was identified in 2002 (77) as a variant of COX-1. This variant is more sensitive to acetaminophen (78). The role of prostanoids in ROP has been demonstrated (73,7981). Other prostanoid compounds, the F-isoprostanes, are generated by lipid peroxidation of AA, independent of COX (82,83). One of these isomers, 8-epi-prostaglandinF (8-epiPGF), is a potent mitogen and constrictor of vascular smooth muscle, and is highly correlated with oxidative stress (83). Studies have demonstrated that 8-epi-PGF is functionally linked to, and activates the thromboxane A2 (TxA2) receptor (84), and may be involved in retinal vasoconstriction and ischemic retinopathies (85). The importance of the prostanoid cascade in ROP and the role of COX-2 in inflammation led to the premise that COX inhibition could have potential benefits. Nonselective COX inhibitors, such as indomethacin and ibuprofen improve OIR and ocular biomarkers of angiogenesis in animal models (8690). However, there are only two clinical studies involving the use of COX inhibitors for prevention of ROP. One controlled study administered topical ketorolac (a non-selective COX inhibitor) to 59 infants (<30 weeks gestation, <1250 grams birth weight) every 8 hours in each eye compared to 53 untreated preterm infants. Ketorolac reduced the risk of developing severe ROP with no significant adverse side effects (91). A later study of 47 preterm infants (<29 weeks, <1000 grams) who receive ketorolac in one eye and placebo in the other eye showed two infants did not develop ROP, 6 showed different ROP staging between the two eyes, 4 of which had a better outcome in the eye receiving ketorolac and no significant treatment-related side effects occurred (92). Dexamethasone is a selective COX-2 inhibitor with anti-inflammatory effects has been demonstrated to reduce the incidence of ROP (93,94). However, its use is associated with significant side effects (95100) and its efficacy in preventing ROP is controversial. Despite overwhelming evidence for the role of COX metabolites in oxidative stress and inflammation, only one prospective randomized, controlled multicenter clinical trial to investigate the benefits of COX inhibitors for prevention of ROP has been initiated and is currently underway (Aranda JV, Clinicaltrials.gov Identifier: NCT02344225).

Inositol

Inositol is a nonglucose carbohydrate that is present in high amounts in the umbilical cord and maternal serum at term (101). Inositol is also present at high concentrations in human breast milk, particularly colostrum suggesting an importance for postnatal growth (102). In 1992, Hallman et al. (103) showed a reduction in ROP in inositol-treated infants (13%) versus placebo (26%) and none of the infants given inositol had stage 4 disease compared to 7 in the placebo group. These findings were later confirmed by Friedman et al. (104). In 2015, a meta-analysis of 4 clinical trials showed reduced stage 3 ROP in 2 trials of the 4 trials (105). A large size multi-center, randomized, controlled trial is currently ongoing to further investigate the benefits of inositol for ROP (Clinicaltrials.gov Identifier: NCT01954082).

Propranolol

Propranolol is one of the newest proposed treatments for ROP despite weak and contradictory animal preclinical data Propranalol is a nonselective β-adrenoreceptor blocker shown to reduce the growth of infantile capillary haemangiomas (106). The mechanism of action is likely due to suppression of VEGF. Studies have shown that polymorphisms of the β-adrenergic receptor in many black infants may be responsible for the lower incidence of ROP (107). Clinical trials have shown reduction in the incidence of ROP with oral propranolol with few side effects (108110). Clinical trials in Israel and Italy are being conducted to evaluate efficacy and safety of this beta blocker for ROP prevention in preterm neonates. (ClinicalTrials.Gov : NCT01238471 ; NCT02014454, NCT02504944,) One Italian Trial using propranolol eye drops of 0.5mg/Kg oral, every 6 hours was discontinued due to serious adverse effects attributed to the pharmacologic actions of propranolol such as severe hypotension, bradycardia or bronchospasm. (Clinical Trials.Gov NCT01079715)

Caffeine

Caffeine is the most widely used drug worldwide (111). It is a methylxanthine that acts to antagonize adenosine receptors and block the release of endogenous adenosine. Caffeine also stimulates dopaminergic activity by removing the negative modulatory effects of adenosine at dopamine receptors (112). Caffeine was first demonstrated to be effective for treatment of apnea of prematurity (AOP) in the 1970s by Aranda et al. (113). After over 40 years, caffeine is now the drug of choice for AOP. It is one of the most commonly prescribed drugs for use in ELGANs (114,115). It is a respiratory stimulant in ELGANs and is effective for reducing the duration of mechanical ventilation (116). The Caffeine for Apnea of Pre- maturity (CAP) trial found that caffeine was also beneficial for reduction the incidence of bronchopulmonary dysplasia (BPD), improving long-term neurodevelopmental outcome, and reducing the incidence of severe ROP (117). The mechanism of caffeine effects on severe ROP remain to be determined, but may involve effects on regulators of angiogenesis such as VEGF, sonic hedgehog matrix metalloproteinases (MMPs), and oxidative stress (118121). Additionally, caffeine potentiates the anti-inflammatory effects of COX inhibitors in activated microglia which may also occur in the retina (122). Caffeine has been used in combination with non-steroidal anti-inflammatory drugs (NSAIDs) for many decades to enhance their analgesic effects, suggesting that caffeine may be an effective adjuvant to NSAIDs (122124). Due to its significant impact on major acute neonatal morbidities including AOP, BPD, PDA, and ROP, caffeine is now recognized as the “wonder drug in neonatology” (125) and the “silver bullet in neonatology” (126). Notwithstanding, only one prospective randomized, controlled multicenter clinical trials to investigate the benefits of caffeine for prevention of ROP has been initiated (Aranda JV, Clinicaltrials.gov Identifier: NCT02344225).

Angiogenic Factors

ROP is characterized by the formation of new and aberrant retinal vasculature, suggesting that angiogenic factors play a major role. The role of angiogenic factors in retinal development was first hypothesized 1948 (127). In 1983, Senger et al. (128) discovered an angiogenic growth factor present in tumor cells which he named vascular permeability factor due to its ability to induce vascular leakage. In 1989, Ferrara and Henzel identified, purified and characterized a heparin-binding angiogenic growth factor specific for vascular endothelial cells which they named VEGF (129). Several other angiogenic factors have been identified, including fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), transforming growth factor (TGF), the angiopoietin family, insulin-like growth factor (IGF), and the Ephrin family, all of which work together to promote functional vascular systems (130). VEGF is a vascular permeability factor. Consistent with this characteristic is its ability to induce the expression of plasminogen activators and MMPs to increase vascular permeability (131,132) which may be the first step in angiogenesis (133). It is now widely accepted that the VEGF family is the most potent inducer of angiogenesis and ROP. Levels of VEGF in the vitreous humor correlate with the degree of neovascularization (134,135). Plasma levels of preterm infants in the first week of life were found to be low (48 pg/mL) compared to term infants (200–450 pg/mL) and adults (10–110 pg/mL) (136). VEGF levels in the aqueous humor of Stage 4 (1,109 pg/mL) and Stage 5 (3520 pg/mL) eyes were considerably higher than control eyes (158 pg/mL) (137). Infants with stage 4 ROP had elevated VEGF levels in the subretinal fluid and decreased levels in Stage 5 (138). Induction of VEGF is also associated with generation of ROS, inflammatory cytokines and prostanoids, possibly via inducible COX-2 (139141). VEGF is now considered the most important target for therapeutic interventions for prevention of severe ROP.

VEGF Inhibitors

Several anti-VEGF drugs have been developed. The first anti-VEGF drug to be approved by the FDA for cancer use was bevacizumab, or Avastin (142). Bevacizumab is a recombinant humanized antibody that binds all forms of VEGF-A. Although it is used off-label, it is not formulated for use within the eye (142). However, off-label use by ophthalmologists demonstrated that Avastin was beneficial when injected into the vitreous of adults with macular degeneration. Later, a truncated form of Avastin called ranibizumab (Lucentis) was FDA approved for ocular use. The first FDA-approved drug for use in ocular neonvascularization was pegaptanib (Macugen). Aflibercept (Eyelea), a human soluble VEGF receptor, which acts as a decoy to trap VEGF, was recently FDA-approved for ocular use. Bevacizumab is the first anti-VEGF drug to be used for treatment of ROP in 2007 (143). Since then, many single center trials and case reports have been published making Bevacizumab, the most widely used drug for treatment of severe ROP. The first meta-analysis of bevacizumab use for ROP was published in 2009 (144). The authors analyzed all case reports, and retrospective and prospective trials in peer-reviewed journals reporting the use of bevacizumab in ROP, including 9 articles, 6 case reports, 1 case series, and 2 retrospective studies of 48 infants administered doses from 0.4 to 1.25 mg and cautioned against potential systemic complications and long-term effects of intravitreal bevacizumab in preterm infants. From 2007 to 2009, all investigators reported favorable outcomes with bevacizumab use in ROP. However, the first report of adverse effects was published in 2010 demonstrating choroidal ruptures at 10 weeks post treatment with laser (145). Other reports of adverse events included vitreous or pre-retinal hemorrhage (146), delayed bilateral retinal detachments at 1 month post treatment (147), and retinal detachment (148). The first multicenter randomized, controlled trial was the BEAT-ROP study which demonstrated an advantage of intravitreal bevacizumab over laser therapy for zone 1 or zone II with stage 3+ ROP by improving structural outcomes, decreasing recurrence, and allowing continued development of peripheral retina (149). In the BEAT-ROP study however, assessments of local and systemic safety profile was not determined and follow-up was only 54 weeks' postmenstrual age. Moshfeghi and Berrocal (150) estimated that 47.7% of recurrences would have occurred after the 54 weeks. This was demonstrated by Hu et al. (151). Studies have since tried to address safety of intravitreal bevacizumab use in ROP and demonstrated numerous acute and latent retinal adverse effects (151168). Interestingly, serum concentrations of bevacizumab concurrent with suppressed systemic VEGF levels lasted up to 60 days post intravitreal administration (169172), and the levels were significantly lower than previously reported in preterm infants (173). These reports demonstrate that bevacizumab escapes from the eye into the systemic circulation and has long-lasting effects on systemic VEGF levels in infants with ROP. A recent meta-analysis of 8 prospective studies of 278 eyes of ROP infants and 15 retrospective studies involving 385 eyes of ROP infants treated with intravitreal bevacizumab showed reoccurrence and acceleration of fibrous traction (174). Other anti-VEGF drugs used in ROP include ranibizumab which has a shorter half-life with the potential for decreased systemic toxicity. Treatment with ranibizumab was also associated with significant adverse outcomes (147,175,176). Combination treatment of intravitreal pegaptanib or bevacizumab injection and laser photocoagulation showed significant benefits in the management of stage 3+ ROP (177,178). Evidence now exist that intravitreal administration of anti-VEGF drugs can have prolonged suppressive effects on systemic VEGF in ELGANs. Considering the role of VEGF in the development of vital organs, more randomized, controlled multicenter trials should be conducted to evaluate dose-response effects and long-term safety profiles in this vulnerable population (179186).

Insulin-like Growth Factor (IGF)-1

Although VEGF plays an important role in the development of ROP, its inhibition does not completely prevent ROP. Another growth factor that is currently being studied for prevention of ROP is IGF-1. IGF-1 influences endothelial cell growth and angiogenesis (187, 188), and plays a critical role in the retinal vasculature development through its interaction with VEGF, acting as a permissive factor for maximum VEGF stimulation of angiogenesis (189,190). Mean IGF-1 level has been shown as significantly and proportionately lower in post-menstrual age matched babies with each stage of ROP than without ROP (191194). Work by Hellstrom and colleagues (191,194) have demonstrated that preterm infants who do not develop ROP have significantly higher circulating levels of IGF-I than those who do. Serum IGF-1 levels to predict ROP has been shown to be a reliable prognostic tool by some studies (195,196), but not others (197199). Only one small single center clinical trial of Premiplex (rhIGF1/rhIGFBP3) pharmacokinetics in 5 preterm infants was conducted. None of the infants developed severe ROP (200). A clinical trial to determine the dose of hIGF-1/rhIGFBP-3 given as a Continuous Infusion, to establish and maintain physiologic levels of serum IGF-1 Levels to prevent ROP is ongoing. (ClinTrials.gov NCT01096784)

Polyunsaturated Fatty Acids (PUFAs)

Omega 6 and Omega-3 PUFAs are essential fatty acids that are important for normal growth, vision and neurodevelopment. Omega-6 PUFAs are formed from linolenic acid (LA) which is converted by desaturation and chain elongation to arachidonic acid (AA), the precursor to eicosanoids (prostaglandins, thromboxane and leukotrienes) (201). Omega-3 PUFAs are formed from alpha-linolenic acid (ALA) which is desaturated and chain elongated to docosahexaenoic acid (DHA) (202). PUFAs are important components of the phospholipid bilayer of cell membranes and contributes to cell structural integrity and function (203). Deficiency of AA and DHA in the membrane alters the physical properties of the lipid bilayer and induces vascular leakage (204). It is well established that omega-3 PUFAs play a key role in the protection against oxidative stress and inflammation. Among the benefits of omega-3 PUFAs, is the ability to regulate eicosanoid metabolism. COX catalyzes the initial oxygenation of PUFAs to produce prostaglandin H (PGH2), a short-lived intermediate, which is further metabolized to prostanoids (205). DHA and its precursor, eicosapentaenoic acid (EPA) reduce the formation of COX-2-derived PGE2 (206), a metabolite of omega-6 PUFA-derived AA. COX-2-derived PGE2 increases VEGF-stimulated invasiveness and angiogenesis through NF-κB (207). There is compelling evidence that EPA could function as a selective COX-2 inhibitor (207209). In the retina, PUFAs accumulate rapidly during the later stages of gestation and early postnatal life (210,211). The retina contains the highest concentration of DHA of all tissues where it is cytoprotective and plays a key role in angiogenic regulation and neuroprotection (212). Administration of fish oil to preterm infants from the first day of life showed a significantly lower incidence and severity of ROP and risk of laser therapy in the treated group (213215). However, two meta-analyses examined the potential benefits of lipid administration and found no significant benefits for the outcome of ROP (216,217). Treatment with omega-3 fatty acids appears to be a promising therapy for prevention and/or treatment of ROP, but there is a paucity of multi-center randomized clinical trials.

DRUGS ASSOCIATED WITH INCIDENCE OF ROP

Surfactant

One of the earlier drugs found to be associated with ROP is surfactant, despite conflicting reports. In the 1990s, there were 3 report of a higher occurrence of ROP with surfactant use (218220). However, comparing the two commonly used surfactants at that time, Exosurf and Survanta, there was a higher occurrence of ROP in the Exosurf group (220). A later meta-analysis involving 10 randomized clinical trials and over 2,000 preterm infants confirmed those findings of a higher risk for ROP with Exosurf (221). Numerous other clinical trials and meta-analyses showed no increased risk for ROP with surfactant therapy.

Erythropoietin (Epo) is routinely used in to treat anemia of prematurity. Epo is a glycoprotein hormone, expressed mainly in the kidney and fetal liver. It is highly responsive to HIF-1α and hypoxia, and is the main regulator of red blood cell production. Many observational and retrospective studies have reported that Epo is associated with an increased risk of ROP (222228), while other studies report no risk (229231). Early meta-analyses involving 23 studies and 2074 preterm infants in 18 countries showed a significant increase in the risk of stage >3 ROP in the Epo group (232). An update in 2014 showed no significant increase in the rate of stage >3 ROP for studies that initiated EPO treatment at less than eight days of age (233). Another meta-analysis done in 2015 also concluded that there were no differences in the incidence of stage >3 ROP with Epo use (234). Clearly, there are major conflicts regarding the role of Epo in the development of ROP, and the reasons for this remain unclear. However, it is well established that both Epo and VEGF are stimulated via HIF-1α during hypoxia, which then leads to angiogenesis (235,238), therefore timing of administration may be crucial.

Steroids

In 1992, two conflicting retrospective studies were published regarding the incidence of ROP requiring laser therapy and postnatal dexamethasone. Batton et al. (237) reported that of the 21 factors examined, only the use of steroids was significantly associated with the need for cryotherapy. On the other hand, Sobel et al. (238) showed a reduction in the need for cryotherapy with prolonged dexamethasone. Several other reports confirmed the findings of Baton et al. (237) of an increased risk of ROP with steroids (239244), while other studies showed no association (245249). However, meta-analyses showed that early exposure to steroids was not associated with a risk of severe ROP (250,251), while exposure at > 7days (252) and >3weeks (253), caused an increase in severe ROP. The use of antenatal steroids was associated with significantly less stage 2 or higher ROP (254,255). Another study comparing antenatal betamethasone with antenatal dexamethasone showed a trend for greater risk of severe ROP associated with dexamethasone (256). Due to its association with increased incidence of cerebral palsy (98), the use of dexamethasone has been restricted.

Indomethacin

Indomethacin is a non-selective COX inhibitor used in neonatology for closure of a patent ductus arteriosus. While studies found no relationship between indomethacin and ROP (257,258), one retrospective study of over 34,000 preterm infants from 162 sites showed a higher incidence of severe ROP (stages 3 and 4) in the group treated with indomethacin on the first day of life (259). Darlow et al. (260) reported that infants treated with indomethacin were 1.5 times more likely to have ROP than untreated infants. Another study involving 105 preterm infants <28 weeks gestation who received low versus high indomethacin showed a significant increase in the incidence of moderate/severe ROP which was directly related to the indomethacin serum concentrations (261). Conversely, a retrospective chart review found that indomethacin use for PDA was protective for ROP (262). It is suggested, that the contradictory effects of indomethacin on ROP may be related to the infant's postnatal age (263). This may indicate that the exact timing of treatment may be crucial and further studies in this regard are needed.

Conclusions

ROP is a multifactorial disease, the cause of which remains largely unknown. Infants at risk suffer from numerous conditions, and are exposed to many forms of polypharmacy which likely add to the etiology of the disease. Since its appearance in the 1940s, pharmacologic interventions for treatment and/or prevention of ROP have predominantly targeted oxidants and VEGF. Furthermore, an overwhelming majority of human and animal studies have focused on rescue treatment of ROP when damage to the retina has already occurred. Given the multi-factorial characteristic of ROP and complexity of the disease, the use of a single therapeutic agent may not be prudent. No one therapy has proven to be effective without adverse effects. The timing of treatment must be considered as it is preferred to prevent any form of ROP rather than minimize its progression. There are conflicting reports regarding the benefits of many of the past and current therapies, most of which were initiated based on anecdotal/observational reports, and in some cases have resulted in severe adverse events to the detriment of the treated infant. Therefore, only data from well-designed, randomized, multi-center, placebo-controlled trials, as well as long-term follow-up studies should be the criteria for implementing a change in clinical practice to use off-label pharmaceuticals in these vulnerable patients.

Acknowledgments

Financial Support: This work was supported by the NIH-Eunice Kennedy Shriver National Institute of Child Health & Human Development Grant #1U54HD071594.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of Interest: The authors have nothing to disclose.

REFERENCES

  • 1.Gilbert C, Fielder A, Gordillo L, et al. Characteristics of infants with severe retinopathy of prematurity in countries with low, moderate, and high levels of development: implications for screening programs. Pediatrics. 2005;115:e518–e525. doi: 10.1542/peds.2004-1180. [DOI] [PubMed] [Google Scholar]
  • 2.Chow LC, Wright KW, Sola A, et al. Can changes in clinical practice decrease the incidence of severe retinopathy of prematurity in very low birth weight infants? Pediatrics. 2003;111:339–345. doi: 10.1542/peds.111.2.339. [DOI] [PubMed] [Google Scholar]
  • 3.Smith L. Pathogenesis of Retinopathy of Prematurity. GHIR. 2004;14:S140–S144. doi: 10.1016/j.ghir.2004.03.030. [DOI] [PubMed] [Google Scholar]
  • 4.Cringle SJ, Yu DY. Oxygen supply and consumption in the retina: implications for studies of retinopathy of prematurity. Doc Ophthalmol. 2010;120:99–109. doi: 10.1007/s10633-009-9197-2. [DOI] [PubMed] [Google Scholar]
  • 5.Kong L, Fry M, Al-Samarraie M, Gilbert C, Steinkuller PG. An update on progress and the changing epidemiology of causes of childhood blindness worldwide. J AAPOS. 2012;16:501–507. doi: 10.1016/j.jaapos.2012.09.004. [DOI] [PubMed] [Google Scholar]
  • 6.York JR, Landers S, Kirby RS, Arbogast PG, Penn JS. Arterial oxygen fluctuation and retinopathy of prematurity in very-low-birth-weight infants. J Perinat. 2004;24:82–87. doi: 10.1038/sj.jp.7211040. [DOI] [PubMed] [Google Scholar]
  • 7.Chan-Ling T, Gock B, Stone J. The effect of oxygen on vasoformative cell division. Evidence that 'physiological hypoxia' is the stimulus for normal retinal vasculogenesis. Invest Ophthalmol Vis Sci. 1995;36:1201–1214. [PubMed] [Google Scholar]
  • 8.Sears NC, Sears JE. Oxygen and retinopathy of prematurity. Int Ophthalmol Clin. 2011;51:17–31. doi: 10.1097/IIO.0b013e3182009916. [DOI] [PubMed] [Google Scholar]
  • 9.Ashton N, Ward B, Serpell G. Effect of oxygen on developing retinal vessels with particular reference to the problem of retrolental fibroplasia. Br J Ophthalmol. 1954;38:397–432. doi: 10.1136/bjo.38.7.397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Raghuveer TS, Bloom BT. A paradigm shift in the prevention of retinopathy of prematurity. Neonatology. 2011;100:116–129. doi: 10.1159/000322848. [DOI] [PubMed] [Google Scholar]
  • 11.Chen J, Smith LE. Retinopathy of prematurity. Angiogenesis. 2007;10:133–140. doi: 10.1007/s10456-007-9066-0. [DOI] [PubMed] [Google Scholar]
  • 12.Di Fiore JM, Bloom JN, Orge F, et al. A higher incidence of intermittent hypoxemic episodes is associated with severe retinopathy of prematurity. J Pediatr. 2010;157:69–73. doi: 10.1016/j.jpeds.2010.01.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Di Fiore JM, Kaffashi F, Loparo K, et al. The relationship between patterns of intermittent hypoxia and retinopathy of prematurity in preterm infants. Pediatr Res. 2012;72:606–612. doi: 10.1038/pr.2012.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Coleman R, Beharry KD, Brock RS, Abad-Santos P, Abad-Santos M, Modanlou HD. Effects of brief clustered versus dispersed hypoxiv episodes on systemic and ocular growth factors in a rat model of OIR. Pediatr Res. 2008;64:50–55. doi: 10.1203/PDR.0b013e31817307ac. [DOI] [PubMed] [Google Scholar]
  • 15.Brock RS, Gebrekristos BH, Kuniyoshi KM, Modanlou HD, Falcao MC, Beharry KD. Biomolecular effects of JB1 (an IGF-I peptide analog) in a rat model of oxygen-induced retinopathy. Pediatr Res. 2011;69:35–41. doi: 10.1203/PDR.0b013e318204e6fa. [DOI] [PubMed] [Google Scholar]
  • 16.Beharry KD, Cai CL, Sharma P, et al. Hydrogen peroxide accumulation in the choroid during intermittent hypoxia increases risk of severe oxygen-induced retinopathy in neonatal rats. IOVS. 2013;54:7644–7657. doi: 10.1167/iovs.13-13040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Jivabhai Patel S, Bany-Mohammed F, McNally L, et al. Exogenous superoxide dismutase mimetic without scavenging H2O2 causes photoreceptor damage in a rat model for oxygen-induced retinopathy. Invest Ophthalmol Vis Sci. 2015;56:1665–1677. doi: 10.1167/iovs.14-15321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hartnett ME. The effects of oxygen stresses on the development of features of severe retinopathy of prematurity: knowledge from the 50/10 OIR model. Doc Ophthalmol. 2010;120:25–39. doi: 10.1007/s10633-009-9181-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.McColm JR, Geisen P, Hartnett ME. VEGF isoforms and their expression after a single episode of hypoxia or repeated fluctuations between hyperoxia and hypoxia: relevance to clinical ROP. Mol Vis. 2004;10:512–520. [PMC free article] [PubMed] [Google Scholar]
  • 20.Tin W, Walker S, Lacamp C. Oxygen monitoring in preterm babies: too high, too low? Paediatr Respir Rev. 2003;4:9–14. doi: 10.1016/s1526-0542(02)00307-x. [DOI] [PubMed] [Google Scholar]
  • 21.Silverman WA. A cautionary tale about supplemental oxygen: the albatross of neonatal medicine. Pediatrics. 2004;113:394–396. doi: 10.1542/peds.113.2.394. [DOI] [PubMed] [Google Scholar]
  • 22.Fleck BW, Stenson BJ. Retinopathy of prematurity and the oxygen conundrum: lessons learned from recent randomized trials. Clin Perinatol. 2013;40:229–240. doi: 10.1016/j.clp.2013.02.010. [DOI] [PubMed] [Google Scholar]
  • 23.Terry TL. Fibroblastic overgrowth of persistent tunica vasculosa lentis in infants born prematurely: II. Report of cases-clinical aspects. Trans Am Ophthalmol Soc. 1942;40:262–284. [PMC free article] [PubMed] [Google Scholar]
  • 24.Campbell K. Intensive oxygen therapy as a possible cause of retrolental fibroplasia; a clinical approach. Med J Aust. 1951;2:48–50. [PubMed] [Google Scholar]
  • 25.Patz A, Hoeck LE, De La Cruz E. Studies on the effect of high oxygen administration in retrolental fibroplasia. I. Nursery observations. Am J Ophthalmol. 1952;35(9):1248–1253. doi: 10.1016/0002-9394(52)91140-9. [DOI] [PubMed] [Google Scholar]
  • 26.Ashton N. Animal experiments in retrolental fibroplasia. Trans Am Acad Ophthalmol Otolaryngol. 1954;58:51–53. [PubMed] [Google Scholar]
  • 27.Bolton DPG, Cross KW. Further observations on the cost of preventing retrolental fibroplasia. Lancet. 1974;1:445–448. doi: 10.1016/s0140-6736(74)92395-2. [DOI] [PubMed] [Google Scholar]
  • 28.STOP-ROP Multicenter Study Group. Supplemental therapeutic oxygen for prethreshold retinopathy of prematurity (STOP-ROP), a randomized, controlled trial. I: primary outcomes. Pediatrics. 2000;105:295–310. doi: 10.1542/peds.105.2.295. [DOI] [PubMed] [Google Scholar]
  • 29.Bancalari E, Claure N. Control of oxygenation during mechanical ventilation in the premature infant. Clin Perinatol. 2012;39:563–572. doi: 10.1016/j.clp.2012.06.013. [DOI] [PubMed] [Google Scholar]
  • 30.Bancalari E, Claure N. Too much or too little: how to handle oxygen saturation in the neonatal intensive care unit. Early Hum Dev. 2012;88(Suppl2):S78–S80. doi: 10.1016/S0378-3782(12)70021-X. [DOI] [PubMed] [Google Scholar]
  • 31.Askie LM, Henderson-Smart DJ, Irwig L, Simpson JM. Oxygen-saturation targets and outcomes in extremely preterm infants. N Engl J Med. 2003;349:959–967. doi: 10.1056/NEJMoa023080. [DOI] [PubMed] [Google Scholar]
  • 32.SUPPORT Study Group of the Eunice Kennedy Shriver NICHD Neonatal Research Network. Target ranges of oxygen saturation in extremely preterm infants. NEJM. 2010;27(362):1959–1969. doi: 10.1056/NEJMoa0911781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.BOOST II United Kingdom Collaborative Group; BOOST II Australia Collaborative Group; BOOST II New Zealand Collaborative Group. Oxygen saturation and outcomes in preterm infants. NEJM. 2013;368:2094–2104. doi: 10.1056/NEJMoa1302298. [DOI] [PubMed] [Google Scholar]
  • 34.Schmidt B, Whyte RK, Asztalos EV, et al. Canadian Oxygen Trial (COT) Group. Effects of targeting higher vs lower arterial oxygen saturations on death or disability in extremely preterm infants: a randomized clinical trial. JAMA. 2013;309:2111–2120. doi: 10.1001/jama.2013.5555. [DOI] [PubMed] [Google Scholar]
  • 35.Manja V, Lakshminrusimha S, Cook DJ. Oxygen saturation target range for extremely preterm infants: a systematic review and meta-analysis. JAMA Pediatr. 2015;169:332–340. doi: 10.1001/jamapediatrics.2014.3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sola A, Golombek SG, Montes Bueno MT, et al. Safe oxygen saturation targeting and monitoring in preterm infants: can we avoid hypoxia and hyperoxia? Acta Paediatr. 2014;103:1009–1018. doi: 10.1111/apa.12692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sorriento D, Pascale AV, Finelli R, et al. Targeting mitochondria as therapeutic strategy for metabolic disorders. Scientific World Journal. 2014;2014:604685. doi: 10.1155/2014/604685. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 38.Weinberger B, Laskin DL, Heck DE, Laskin JD. Oxygen toxicity in premature infants. Toxicol Appl Pharmacol. 2002;181:60–67. doi: 10.1006/taap.2002.9387. [DOI] [PubMed] [Google Scholar]
  • 39.Katz ML, Robison WG., Jr Autoxidative damage to the retina: Potential role in retinopathy of prematurity. Birth Defects. 1988;24:237–248. [PubMed] [Google Scholar]
  • 40.Hardy P, Beauchamp M, Sennlaub F, et al. Inflammatory lipid mediators in ischemic retinopathy. Pharmacol Rep. 2005;57(Suppl):169–190. [PubMed] [Google Scholar]
  • 41.McElroy MC, Postle AD, Kelly FJ. Catalase, superoxide dismutase and glutathione peroxidase activities of lung and liver during human development. Biochim Biophys Acta. 1992;1117:153–158. doi: 10.1016/0304-4165(92)90073-4. [DOI] [PubMed] [Google Scholar]
  • 42.Lindeman JH1, van Zoeren-Grobben D, Schrijver J, Speek AJ, Poorthuis BJ, Berger HM. The total free radical trapping ability of cord blood plasma in preterm and term babies. Pediatr Res. 1989;26:20–24. doi: 10.1203/00006450-198907000-00008. [DOI] [PubMed] [Google Scholar]
  • 43.Sullivan JL1, Newton RB. Serum antioxidant activity in neonates. Arch Dis Child. 1988;63:748–750. doi: 10.1136/adc.63.7_spec_no.748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Owens WC, Owens EU. Retrolental fibroplasia in premature infants. II. Studies on the prophylaxis of the disease: the use of alpha tocopheryl acetate. Am J Ophthalmol. 1949;32:1631–1637. [PubMed] [Google Scholar]
  • 45.Johnson L, Schaffer D, Quinn G, et al. Vitamin E supplementation and the retinopathy of prematurity. Ann N Y Acad Sci. 1982;393:473–495. doi: 10.1111/j.1749-6632.1982.tb31285.x. [DOI] [PubMed] [Google Scholar]
  • 46.Johnson L, Abbasi S, Quinn GE, Otis C, Bowen FW., Jr Vitamin E and retinopathy of prematurity. Pediatrics. 1988;81:329–331. [PubMed] [Google Scholar]
  • 47.Johnson L, Quinn GE, Abbasi S, et al. Effect of sustained pharmacologic vitamin E levels on incidence and severity of retinopathy of prematurity: a controlled clinical trial. J Pediatr. 1989;114:827–838. doi: 10.1016/s0022-3476(89)80149-0. [DOI] [PubMed] [Google Scholar]
  • 48.Finer NN, Schindler RF, Grant G, Hill GB, Peters K. Effect of intramuscular vitamin E on frequency and severity of retrolental fibroplasia. A controlled trial. Lancet. 1982;1:1087–1091. doi: 10.1016/s0140-6736(82)92276-0. [DOI] [PubMed] [Google Scholar]
  • 49.Schaffer DB, Johnson L, Quinn GE, Weston M, Bowen FW., Jr Vitamin E and retinopathy of prematurity. Follow-up at one year. Ophthalmology. 1985;92:1005–1011. doi: 10.1016/s0161-6420(85)33913-1. [DOI] [PubMed] [Google Scholar]
  • 50.Puklin JE, Simon RM, Ehrenkranz RA. Influence on retrolental fibroplasia of intramuscular vitamin E administration during respiratory distress syndrome. Ophthalmology. 1982;89:96–103. doi: 10.1016/s0161-6420(82)34830-7. [DOI] [PubMed] [Google Scholar]
  • 51.Hittner HM, Godio LB, Rudolph AJ, et al. Retrolental fibroplasia: efficacy of vitamin E in a double-blind clinical study of preterm infants. NEJM. 1981;305:1365–1371. doi: 10.1056/NEJM198112033052301. [DOI] [PubMed] [Google Scholar]
  • 52.Phelps DL, Rosenbaum AL, Isenberg SJ, Leake RD, Dorey FJ. Tocopherol efficacy and safety for preventing retinopathy of prematurity: a randomized, controlled, double-masked trial. Pediatrics. 1987;79:489–500. [PubMed] [Google Scholar]
  • 53.Rosenbaum AL, Phelps DL, Isenberg SJ, Leake RD, Dorey F. Retinal hemorrhage in retinopathy of prematurity associated with tocopherol treatment. Ophthalmology. 1985;92:1012–1014. doi: 10.1016/s0161-6420(85)33912-x. [DOI] [PubMed] [Google Scholar]
  • 54.Raju TN, Langenberg P, Bhutani V, Quinn GE. Vitamin E prophylaxis to reduce retinopathy of prematurity: a reappraisal of published trials. J Pediatr. 1997;131:844–850. doi: 10.1016/s0022-3476(97)70031-3. [DOI] [PubMed] [Google Scholar]
  • 55.Brion LP, Bell EF, Raghuveer TS. Vitamin E supplementation for prevention of morbidity and mortality in preterm infants. Cochrane Database Syst Rev. 2003:CD003665. doi: 10.1002/14651858.CD003665. [DOI] [PubMed] [Google Scholar]
  • 56.Law MR, Wijewardene K, Wald NJ. Is routine vitamin E administration justified in very low-birthweight infants? Dev Med Child Neurol. 1990;32:442–450. doi: 10.1111/j.1469-8749.1990.tb16963.x. [DOI] [PubMed] [Google Scholar]
  • 57.Christensen RD, Alder SC, Richards SC, Horn JT, Lambert DK, Baer VL. A pilot trial testing the feasibility of administering D-penicillamine to extremely low birth weight neonates. J Perinatol. 2006;26:120–124. doi: 10.1038/sj.jp.7211440. [DOI] [PubMed] [Google Scholar]
  • 58.Lakatos L, Hatvani I, Oroszlán G, et al. Controlled trial of D-penicillamine to prevent retinopathy of prematurity. Acta Paediatr Hung. 1986;27:47–56. [PubMed] [Google Scholar]
  • 59.Vekerdy-Lakatos S, Lakatos L, Oroszlán G, Itzés B. One year longitudinal follow-up of premature infants treated with D-penicillamine in the neonatal period. Acta Paediatr Hung. 1987;28:9–16. [PubMed] [Google Scholar]
  • 60.Tandon M1, Dutta S, Dogra MR, Gupta A. Oral D-penicillamine for the prevention of retinopathy of prematurity in very low birth weight infants: a randomized, placebo-controlled trial. Acta Paediatr. 2010;99:1324–1328. doi: 10.1111/j.1651-2227.2010.01837.x. [DOI] [PubMed] [Google Scholar]
  • 61.Qureshi MJ, Kumar M. D-Penicillamine for preventing retinopathy of prematurity in preterm infants. Cochrane Database Syst Rev. 2013;9:CD001073. doi: 10.1002/14651858.CD001073.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Parad RB, Allred EN, Rosenfeld WN, Davis JM. Reduction of retinopathy of prematurity in extremely low gestational age newborns treated with recombinant human Cu/Zn superoxide dismutase. Neonatology. 2012;102:139–144. doi: 10.1159/000336639. [DOI] [PubMed] [Google Scholar]
  • 63.Romagnoli C1, Giannantonio C, Cota F, et al. A prospective, randomized, double blind study comparing lutein to placebo for reducing occurrence and severity of retinopathy of prematurity. J Matern Fetal Neonatal Med. 2011;24(Suppl 1):147–150. doi: 10.3109/14767058.2011.607618. [DOI] [PubMed] [Google Scholar]
  • 64.Dani C, Lori I, Favelli F, et al. Lutein and zeaxanthin supplementation in preterm infants to prevent retinopathy of prematurity: a randomized controlled study. J Matern Fetal Neonatal Med. 2012;25:523–527. doi: 10.3109/14767058.2011.629252. [DOI] [PubMed] [Google Scholar]
  • 65.Manzoni P1, Guardione R, Bonetti P, Priolo C, Maestri A, Mansoldo C, Mostert M, Anselmetti G, Sardei D, Bellettato M, Biban P, Farina D. Lutein and zeaxanthin supplementation in preterm very low-birth-weight neonates in neonatal intensive care units: a multicenter randomized controlled trial. Am J Perinatol. 2013 Jan;30(1):25–32. doi: 10.1055/s-0032-1321494. [DOI] [PubMed] [Google Scholar]
  • 66.Hopkins FG. Feeding experiments illustrating the importance of accessory factors in normal dietaries. J Physiol. 1912;44:425–460. doi: 10.1113/jphysiol.1912.sp001524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mactier H, Weaver LT. Vitamin A and preterm infants: what we know, what we don't know, and what we need to know. Arch Dis Child Fetal Neonatal Ed. 2005;90:F103–F108. doi: 10.1136/adc.2004.057547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mactier H, McCulloch DL, Hamilton R, et al. Vitamin A supplementation improves retinal function in infants at risk of retinopathy of prematurity. J Pediatr. 2012;160:954–959. doi: 10.1016/j.jpeds.2011.12.013. [DOI] [PubMed] [Google Scholar]
  • 69.Ambalavanan N, Wu TJ, Tyson JE, Kennedy KA, Roane C, Carlo WA. A comparison of three vitamin A dosing regimens in extremely-low-birth-weight infants. J Pediatr. 2003;142:656–661. doi: 10.1067/mpd.2003.214. [DOI] [PubMed] [Google Scholar]
  • 70.Darlow BA, Graham PJ. Vitamin A supplementation for preventing morbidity and mortality in very low birthweight infants. Cochrane Database Syst Rev. 2002;(4):CD000501. doi: 10.1002/14651858.CD000501. [DOI] [PubMed] [Google Scholar]
  • 71.Russell GA, Cooke RW. Randomised controlled trial of allopurinol prophylaxis in very preterm infants. Arch Dis Child Fetal Neonatal Ed. 1995;73:F27–F31. doi: 10.1136/fn.73.1.f27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Korbecki J, Baranowska-Bosiacka I, Gutowska I, Chlubek D. The effect of reactive oxygen species on the synthesis of prostanoids from arachidonic acid. J Physiol Pharmacol. 2013;64:409–421. [PubMed] [Google Scholar]
  • 73.Hardy P, Dumont I, Bhattacharya M, et al. Oxidants, nitric oxide and prostanoids in the developing ocular vasculature: a basis for ischemic retinopathy. Cardiovasc Res. 2000;47:489–509. doi: 10.1016/s0008-6363(00)00084-5. [DOI] [PubMed] [Google Scholar]
  • 74.Balsinde J, Winstead MV, Dennis EA. Phospholipase A(2) regulation of arachidonic acid mobilization. FEBS Lett. 2002;531:2–6. doi: 10.1016/s0014-5793(02)03413-0. [DOI] [PubMed] [Google Scholar]
  • 75.Gilroy DW, Tomlinson A, Willoughby A. Differential effects of inhibition of isoforms of cyclooxygenase (COX-1, COX-2) in chronic inflammation. Inflamm Res. 1998;47:79–85. doi: 10.1007/s000110050285. [DOI] [PubMed] [Google Scholar]
  • 76.FitzGerald GA, Patrono C. The coxibs, selective inhibitors of cyclooxygenase-2. NEJM. 2001;345:433–442. doi: 10.1056/NEJM200108093450607. [DOI] [PubMed] [Google Scholar]
  • 77.Chandrasekharan NV, Dai H, Roos KL, et al. COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: cloning, structure, and expression. Proc Natl Acad Sci USA. 2002;99:13926–13931. doi: 10.1073/pnas.162468699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Botting RM. Vane's discovery of the mechanism of action of aspirin changed our understanding of its clinical pharmacology. Pharmacol Rep. 2010;62:518–525. doi: 10.1016/s1734-1140(10)70308-x. [DOI] [PubMed] [Google Scholar]
  • 79.Stuart MJ, Phelps DL, Setty BN. Changes in oxygen tension and effects on cyclooxygenase metabolites: II. Decrease of retinal prostacyclin in kittens exposed to hyperoxia. Pediatrics. 1988;82:367–372. [PubMed] [Google Scholar]
  • 80.Stuart MJ, Setty Y, Walenga RW, Graeber JE, Ganley C. Effects of hyperoxia and hypoxia on vascular prostacyclin formation in vitro. Pediatrics. 1984;74:548–553. [PubMed] [Google Scholar]
  • 81.Cadell JL. Hypothesis: the possible role of magnesium and copper deficiency in retinopathy of prematurity. Magnes Res. 1995;8:261–270. [PubMed] [Google Scholar]
  • 82.Morrow JD, Scruggs J, Chen Y, Zackert WE, Roberts LJ., II Evidence that the E2-is isoprostane, 15-E2t-isoprostane (8-iso-prostaglandin E2) is formed in vivo. J Lipid Res. 1998;39:1589–1593. [PubMed] [Google Scholar]
  • 83.Kawikova I, Barnes PJ, Takahashi T, Tadjkarimi S, Yacoub MH, Belvisi MG. 8-epi-PGF2α, a novel noncyclooxygenase-derived prostaglandin, constricts airways in vitro. Am J Respir Crit Care Med. 1996;153:590–596. doi: 10.1164/ajrccm.153.2.8564103. [DOI] [PubMed] [Google Scholar]
  • 84.Kinsella BT, O’Mahony DJ, Fitzgerald GA. The human thromboxane A2 receptor alpha isoform (TP alpha) functionally couples to the G proteins Gq and G11 in vivo and is activated by the isoprostane 8-epi prostaglandin F2 alpha. J Pharmacol Exp Ther. 1997;281:957–964. [PubMed] [Google Scholar]
  • 85.Lahaie I, Hardy P, Hou X, et al. A novel mechanism for vasoconstrictor action of 8-isoprostaglandin F2 alpha on retinal vessels. Am J Physiol. 1998;274:R1406–R1416. doi: 10.1152/ajpregu.1998.274.5.R1406. [DOI] [PubMed] [Google Scholar]
  • 86.Chemtob S, Beharry K, Rex J, Chatterjee T, Varma DR, Aranda JV. Ibuprofen enhances retinal and choroidal blood flow autoregulation in newborn piglets. Invest Ophthalmol Vis Sci. 1991;32:1799–1807. [PubMed] [Google Scholar]
  • 87.Sakamoto T1, Soriano D, Nassaralla J, et al. Effect of intravitreal administration of indomethacin on experimental subretinal neovascularizationin the subhuman primate. Arch Ophthalmol. 1995;113:222–226. doi: 10.1001/archopht.1995.01100020106040. [DOI] [PubMed] [Google Scholar]
  • 88.Nandgaonkar BN, Rotschild TUK, Higgins RD. Indomethacin improves oxygen-induced retinopathy of prematurity in the mouse. Pediatr Res. 1999;46:184–188. doi: 10.1203/00006450-199908000-00010. [DOI] [PubMed] [Google Scholar]
  • 89.Sharma J, Barr SM, Geng Y, Yun Y, Higgins RD. Ibuprofen improves oxygen-induced retinopathy in a mouse model. Curr Eye Res. 2003;27:309–314. doi: 10.1076/ceyr.27.5.309.17222. [DOI] [PubMed] [Google Scholar]
  • 90.Beharry KD1, Modanlou HD, Hasan J, et al. Comparative effects of early postnatal ibuprofen and indomethacin on VEGF, IGF-I, and GH during rat ocular development. Invest Ophthalmol Vis Sci. 2006;47:3036–3043. doi: 10.1167/iovs.06-0057. [DOI] [PubMed] [Google Scholar]
  • 91.Avila-Vazquez M, Maffrand R, Sosa M, et al. Treatment of retinopathy of prematurity with topical ketorolac tromethamine: a preliminary study. BMC Pediatr. 2004;7:4–15. doi: 10.1186/1471-2431-4-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Giannantonio C, Purcaro V, Cota F, et al. Effectiveness of ketorolac tromethamine in prevention of severe retinopathy of prematurity. J Pediatr Ophthalmol Strabismus. 2010:1–5. doi: 10.3928/01913913-20100920-01. [DOI] [PubMed] [Google Scholar]
  • 93.Higgins RD, Mendelsohn AL, DeFeo MJ, Ucsel R, Hendricks-Munoz KD. Antenatal dexamethasone and decreased severity of retinopathy of prematurity. Arch Ophthalmol. 1998;116:601–605. doi: 10.1001/archopht.116.5.601. [DOI] [PubMed] [Google Scholar]
  • 94.Doyle LW, Ehrenkranz RA, Halliday HL. Dexamethasone treatment in the first week of life for preventing bronchopulmonary dysplasia in preterm infants: a systematic review. Neonatology. 2010;98:217–224. doi: 10.1159/000286210. [DOI] [PubMed] [Google Scholar]
  • 95.Modanlou HD, Beharry K. Combined effects of dexamethasone and surfactant supplementation on outcome of VLBW infants. J Perinatol. 1996;16:422–428. [PubMed] [Google Scholar]
  • 96.Smith LM, Qureshi N, Chao CR. Effects of single and multiple courses of antenatal glucocorticoids in preterm newborn less than 30 weeks’ gestation. J Matern Fetal Med. 2000;9:131–135. doi: 10.1002/(SICI)1520-6661(200003/04)9:2<131::AID-MFM9>3.0.CO;2-M. [DOI] [PubMed] [Google Scholar]
  • 97.National Institutes of Child Health and Human Development Neonatal Research Network. Adverse effects of early dexamethasone treatment in extremely-low-birth-weight infants. NEJM. 2001;344:95–101. doi: 10.1056/NEJM200101113440203. [DOI] [PubMed] [Google Scholar]
  • 98.Yeh TF, Lin YJ, Huang CC, et al. Early dexamethasone therapy in preterm infants: a followup study. Pediatrics. 1998;101:E7. doi: 10.1542/peds.101.5.e7. [DOI] [PubMed] [Google Scholar]
  • 99.O’Shea TM, Jacks KE, Klinepeter KL, Peters NJ, Dillard RG. Antenatal betamethasone and the risk of cerebral palsy (CP) in very low birth weight (VLBW) infants. Pediatric Res. 1999;45:252A. [Google Scholar]
  • 100.Bakker JM, Kavelaars A, Kamphuis PJGH, Heijnen CJ, van Bel F. Effects of early neonatal dexamethasone treatment on susceptibility to experimental autoimmune encephalomyelitis (EAE) and on the LPS-induced corticosterone responses in adult rats. Pediatr Res. 1999;45:48A. [Google Scholar]
  • 101.Brusati V, Józwik M, Józwik M, et al. Fetal and maternal non-glucose carbohydrates and polyols concentrations in normal human pregnancies at term. Pediatr Res. 2005;58:700–704. doi: 10.1203/01.PDR.0000180549.86614.73. [DOI] [PubMed] [Google Scholar]
  • 102.Cavalli C, Teng C, Battaglia FC, Bevilacqua G. Free sugar and sugar alcohol concentrations in human breast milk. J Pediatr Gastroenterol Nutr. 2006;42:215–221. doi: 10.1097/01.mpg.0000189341.38634.77. [DOI] [PubMed] [Google Scholar]
  • 103.Hallman M, Bry K, Hoppu K, Lappi M, Pohjavuori M. Inositol supplementation in premature infants with respiratory distress syndrome. NEJM. 1992;326:1233–1239. doi: 10.1056/NEJM199205073261901. [DOI] [PubMed] [Google Scholar]
  • 104.Friedman CA, McVey J, Borne MJ, et al. Relationship between serum inositol concentration and development of retinopathy of prematurity: a prospective study. J Pediatr Ophthalmol Strabismus. 2000;37:79–86. doi: 10.3928/0191-3913-20000301-06. [DOI] [PubMed] [Google Scholar]
  • 105.Howlett A, Ohlsson A, Plakkal N. Inositol in preterm infants at risk for or having respiratory distress syndrome. Cochrane Database Syst Rev. 2015;2:CD000366. doi: 10.1002/14651858.CD000366.pub3. [DOI] [PubMed] [Google Scholar]
  • 106.Léauté-Labrèze C. Propranolol in infantile hemangiomas. Arch Pediatr. 2015;22:452–455. doi: 10.1016/j.arcped.2015.01.001. [DOI] [PubMed] [Google Scholar]
  • 107.Palmer EA, Flynn JT, Hardy RJ, et al. Incidence and early course of retinopathy of prematurity. The Cryotherapy for Retinopathy of Prematurity Cooperative Group. Ophthalmology. 1991;98:1628–1640. doi: 10.1016/s0161-6420(91)32074-8. [DOI] [PubMed] [Google Scholar]
  • 108.Filippi L, Cavallaro G, Bagnoli P, et al. Oral propranolol for retinopathy of prematurity: risks, safety concerns, and perspectives. J Pediatr. 2013;163:1570–1577. doi: 10.1016/j.jpeds.2013.07.049. [DOI] [PubMed] [Google Scholar]
  • 109.Bührer C, Bassler D. Oral Propranolol: A New Treatment for Infants with Retinopathy of Prematurity? Neonatology. 2015;108:49–52. doi: 10.1159/000381659. [DOI] [PubMed] [Google Scholar]
  • 110.Makhoul IR, Peleg O, et al. Oral propranolol versus placebo for retinopathy of prematurity: a pilot, randomised, double-blind prospective study. Arch Dis Child. 2013;98:565–567. doi: 10.1136/archdischild-2013-303951. [DOI] [PubMed] [Google Scholar]
  • 111.Meredith SE, Juliano LM, Hughes JR, Griffiths RR. Caffeine Use Disorder: A Comprehensive Review and Research Agenda. J Caffeine Res. 2013;3:114–130. doi: 10.1089/jcr.2013.0016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ferré S. An update on the mechanisms of the psychostimulant effects of caffeine. J Neurochem. 2008;105:1067–1079. doi: 10.1111/j.1471-4159.2007.05196.x. [DOI] [PubMed] [Google Scholar]
  • 113.Aranda JV, Gorman W, Bergsteinsson H. Efficacy of caffeine in treatment of apnea in the low-birth-weight infant. J Pediatr. 1977;90:467–472. doi: 10.1016/s0022-3476(77)80718-x. [DOI] [PubMed] [Google Scholar]
  • 114.Kreutzer K, Bassler D. Caffeine for apnea of prematurity: a neonatal success story. Neonatology. 2014;105:332–336. doi: 10.1159/000360647. [DOI] [PubMed] [Google Scholar]
  • 115.Bancalari E. Caffeine for apnea of prematurity. NEJM. 2006;354:2179–2181. doi: 10.1056/NEJMe068028. [DOI] [PubMed] [Google Scholar]
  • 116.Steer P, Flenady V, Shearman A, et al. Caffeine Collaborative Study Group Steering Group. High dose caffeine citrate for extubation of preterm infants: a randomised controlled trial. Arch Dis Child Fetal Neonatal Ed. 2004;89:F499–F503. doi: 10.1136/adc.2002.023432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Schmidt B1, Roberts RS, Davis P, et al. Caffeine for Apnea of Prematurity Trial Group. Long-term effects of caffeine therapy for apnea of prematurity. NEJM. 2007;357:1893–1902. doi: 10.1056/NEJMoa073679. [DOI] [PubMed] [Google Scholar]
  • 118.Hsu SJ1, Lee FY, Wang SS, et al. Caffeine ameliorates hemodynamic derangements and portosystemic collaterals in cirrhotic rats. Hepatology. 2015;61:1672–1684. doi: 10.1002/hep.27679. [DOI] [PubMed] [Google Scholar]
  • 119.Sahir N1, Evrard P, Gressens P. Caffeine induces sonic hedgehog gene expression in cultured astrocytes and neurons. J Mol Neurosci. 2004;24:201–205. doi: 10.1385/JMN:24:2:201. [DOI] [PubMed] [Google Scholar]
  • 120.Liu WH1, Chang LS. Caffeine induces matrix metalloproteinase-2 (MMP-2) and MMP-9 down-regulation in human leukemia U937 cells via Ca2+/ROS-mediated suppression of ERK/c-fos pathway and activation of p38 MAPK/c-jun pathway. J Cell Physiol. 2010;224:775–785. doi: 10.1002/jcp.22180. [DOI] [PubMed] [Google Scholar]
  • 121.Varma SD1, Hegde KR, Kovtun S. Oxidative stress in lens in vivo: inhibitory effect of caffeine. A preliminary report. Mol Vis. 2010;16:501–505. [PMC free article] [PubMed] [Google Scholar]
  • 122.Fiebich BL, Lieb K, Hüll M, et al. Effects of caffeine and paracetamol alone or in combination with acetylsalicylic acid on prostaglandin E(2) synthesis in rat microglial cells. Neuropharmacology. 2000;39:2205–2213. doi: 10.1016/s0028-3908(00)00045-9. [DOI] [PubMed] [Google Scholar]
  • 123.Dooley JM, Gordon KE, Wood EP, Brna PM, MacSween J, Fraser A. Caffeine as an adjuvant to ibuprofen in treating childhood headaches. Pediatr Neurol. 2007;37:42–46. doi: 10.1016/j.pediatrneurol.2007.02.016. [DOI] [PubMed] [Google Scholar]
  • 124.López JR, Domínguez-Ramírez AM, Cook HJ, et al. Enhancement of antinociception by co-administration of ibuprofen and caffeine in arthritic rats. Eur J Pharmacol. 2006;544:31–38. doi: 10.1016/j.ejphar.2006.06.041. [DOI] [PubMed] [Google Scholar]
  • 125.Vento M, Curstedt T, Halliday HL, Hallman M, Saugstad OD, Speer CP. More about surfactant, oxygen, caffeine and chronic lung disease. Neonatology. 2014;105:320–322. doi: 10.1159/000360653. [DOI] [PubMed] [Google Scholar]
  • 126.Aranda JV, Beharry K, Valencia GB, Natarajan G, Davis J. Caffeine impact on neonatal morbidities. J Matern Fetal Neonatal Med. 2010;23(Suppl 3):20–23. doi: 10.3109/14767058.2010.517704. [DOI] [PubMed] [Google Scholar]
  • 127.Michaelson IC. The mode of development of the vascular system of the retina, with some observations on its significance for certain retinal disease. Trans Ophthalmol Soc UK. 1948;68:137–180. [Google Scholar]
  • 128.Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 1983;219:983–985. doi: 10.1126/science.6823562. [DOI] [PubMed] [Google Scholar]
  • 129.Ferrara N1, Henzel WJ. Pituitary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cells. Biochem Biophys Res Commun. 1989;161:851–858. doi: 10.1016/0006-291x(89)92678-8. [DOI] [PubMed] [Google Scholar]
  • 130.Gale NW1, Yancopoulos GD. Growth factors acting via endothelial cell-specific receptor tyrosine kinases: VEGFs, angiopoietins, and ephrins in vascular development. Genes Dev. 1999;13:1055–1066. doi: 10.1101/gad.13.9.1055. [DOI] [PubMed] [Google Scholar]
  • 131.Pepper MS1, Ferrara N, Orci L, Montesano R. Vascular endothelial growth factor (VEGF) induces plasminogen activators and plasminogen activator inhibitor-1 in microvascular endothelial cells. Biochem Biophys Res Commun. 1991;181:902–906. doi: 10.1016/0006-291x(91)91276-i. [DOI] [PubMed] [Google Scholar]
  • 132.Unemori EN, Ferrara N, Bauer EA, Amento EP. Vascular endothelial growth factor induces interstitial collagenase expression in human endothelial cells. J Cell Physiol. 1992;153:557–562. doi: 10.1002/jcp.1041530317. [DOI] [PubMed] [Google Scholar]
  • 133.Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. NEJM. 1986;315:1650–1659. doi: 10.1056/NEJM198612253152606. [DOI] [PubMed] [Google Scholar]
  • 134.Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders. NEJM. 1994;331:1480–1487. doi: 10.1056/NEJM199412013312203. [DOI] [PubMed] [Google Scholar]
  • 135.Donahue ML, Phelps DL, Watkins RH, LoMonaco MB, Horowitz S. Retinal vascular endothelial growth factor (VEGF) mRNA expression is altered in relation to neovascularization in oxygen induced retinopathy. Current Eye Res. 1996;15:175–184. doi: 10.3109/02713689608997411. [DOI] [PubMed] [Google Scholar]
  • 136.Lassus P, Ristimäki A, Ylikorkala O, Viinikka L, Andersson S. Vascular endothelial growth factor in human preterm lung. Am J Respir Crit Care Med. 1999;159:1429–1433. doi: 10.1164/ajrccm.159.5.9806073. [DOI] [PubMed] [Google Scholar]
  • 137.Nonobe NI1, Kachi S, Kondo M, et al. Concentration of vascular endothelial growth factor in aqueous humor of eyes with advanced retinopathy of prematurity before and after intravitreal injection of bevacizumab. Retina. 2009;29:579–585. doi: 10.1097/IAE.0b013e3181a3b848. [DOI] [PubMed] [Google Scholar]
  • 138.Lashkari K1, Hirose T, Yazdany J, McMeel JW, Kazlauskas A, Rahimi N. Vascular endothelial growth factor and hepatocyte growth factor levels are differentially elevated in patients with advanced retinopathy of prematurity. Am J Pathol. 2000;156:1337–1344. doi: 10.1016/S0002-9440(10)65004-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Kuroki M, Voest EE, Amano S. Reactive oxygen intermediates increase vascular endothelial growth factor expression in vitro and in vivo. J Clin Invest. 1996;98:1667–1675. doi: 10.1172/JCI118962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ben-Av P, Crofford LJ, Wilder RL, Hla T. Induction of vascular endothelial growth factor expression in synovial fibroblasts by prostaglandin E and interleukin-1: a potential mechanism for inflammatory angiogenesis. FEBS Lett. 1995;372:83–87. doi: 10.1016/0014-5793(95)00956-a. [DOI] [PubMed] [Google Scholar]
  • 141.Lukiw WJ1, Ottlecz A, Lambrou G, et al. Coordinate activation of HIF-1 and NF-kappaB DNA binding and COX-2 and VEGF expression in retinal cells by hypoxia. Invest Ophthalmol Vis Sci. 2003;44:4163–4170. doi: 10.1167/iovs.02-0655. [DOI] [PubMed] [Google Scholar]
  • 142.Tah V, Orlans HO, Hyer J, et al. Anti-VEGF Therapy and the Retina: An Update. J Ophthalmol. 2015;2015:627674. doi: 10.1155/2015/627674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Shah PK, Narendran V, Tawansy KA, Raghuram A, Narendran K. Intravitreal bevacizumab (Avastin) for post laser anterior segment ischemia in aggressive posterior retinopathy of prematurity. Indian J Ophthalmol. 2007;55:75–76. doi: 10.4103/0301-4738.29505. [DOI] [PubMed] [Google Scholar]
  • 144.Micieli JA, Surkont M, Smith AF. A systematic analysis of the off-label use of bevacizumab for severe retinopathy of prematurity. Am J Ophthalmol. 2009;148:536–543. doi: 10.1016/j.ajo.2009.05.031. [DOI] [PubMed] [Google Scholar]
  • 145.Atchaneeyasakul LO, Trinavarat A. Choroidal ruptures after adjuvant intravitreal injection of bevacizumab for aggressive posterior retinopathy of prematurity. J Perinatol. 2010;30:497–499. doi: 10.1038/jp.2009.166. [DOI] [PubMed] [Google Scholar]
  • 146.Wu WC, Yeh PT, Chen SN, Yang CM, Lai CC, Kuo HK. Effects and complications of bevacizumab use in patients with retinopathy of prematurity: a multicenter study in Taiwan. Ophthalmology. 2011;118:176–183. doi: 10.1016/j.ophtha.2010.04.018. [DOI] [PubMed] [Google Scholar]
  • 147.Jang SY, Choi KS, Lee SJ. Delayed-onset retinal detachment after an intravitreal injection of ranibizumab for zone 1 plus retinopathy of prematurity. J AAPOS. 2010;14:457–459. doi: 10.1016/j.jaapos.2010.05.011. [DOI] [PubMed] [Google Scholar]
  • 148.Suk KK, Berrocal AM, Murray TG, et al. Retinal detachment despite aggressive management of aggressive posterior retinopathy of prematurity. J Pediatr Ophthalmol Strabismus. 2010;47:e1–e4. doi: 10.3928/01913913-20101217-06. [DOI] [PubMed] [Google Scholar]
  • 149.Mintz-Hittner HA, Kennedy KA, Chuang AZ BEAT-ROP Cooperative Group. Efficacy of intravitreal bevacizumab for stage 3+ retinopathy of prematurity. NEJM. 2011;364:603–615. doi: 10.1056/NEJMoa1007374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Moshfeghi DM, Berrocal AM. Retinopathy of prematurity in the time of bevacizumab: incorporating the BEAT-ROP results into clinical practice. Ophthalmology. 2011;118:1227–1228. doi: 10.1016/j.ophtha.2011.04.028. [DOI] [PubMed] [Google Scholar]
  • 151.Hu J, Blair MP, Shapiro MJ, Lichtenstein SJ, Galasso JM, Kapur R. Reactivation of retinopathy of prematurity after bevacizumab injection. Arch Ophthalmol. 2012;130:1000–1006. doi: 10.1001/archophthalmol.2012.592. [DOI] [PubMed] [Google Scholar]
  • 152.Henaine-Berra A, Garcia-Aguirre G, Quiroz-Mercado H, Martinez-Castellanos MA. Retinal fluorescein angiographic changes following intravitreal anti-VEGF therapy. J AAPOS. 2014;18:120–123. doi: 10.1016/j.jaapos.2013.12.009. [DOI] [PubMed] [Google Scholar]
  • 153.Ittiara S, Blair MP, Shapiro MJ, Lichtenstein SJ. Exudative retinopathy and detachment: a late reactivation of retinopathy of prematurity after intravitrealbevacizumab. J AAPOS. 2013;17:323–325. doi: 10.1016/j.jaapos.2013.01.004. [DOI] [PubMed] [Google Scholar]
  • 154.Jalali S, Balakrishnan D, Zeynalova Z, Padhi TR, Rani PK. Serious adverse events and visual outcomes of rescue therapy using adjunct bevacizumab to laser and surgery for retinopathy of prematurity. The Indian Twin Cities Retinopathy of Prematurity Screening database Report number 5. Arch Dis Child Fetal Neonatal Ed. 2012;98:F327–F333. doi: 10.1136/archdischild-2012-302365. [DOI] [PubMed] [Google Scholar]
  • 155.Karaca C, Oner AO, Mirza E, Polat OA, Sahiner M. Bilateral effect of unilateral bevacizumab injection in retinopathy of prematurity. JAMA Ophthalmol. 2013;131:1099–1101. doi: 10.1001/jamaophthalmol.2013.400. [DOI] [PubMed] [Google Scholar]
  • 156.Martínez-Castellanos MA, Schwartz S, Hernández-Rojas ML, et al. Long-term effect of antiangiogenic therapy for retinopathy of prematurity up to 5 years of follow-up. Retina. 2013;33:329–338. doi: 10.1097/IAE.0b013e318275394a. [DOI] [PubMed] [Google Scholar]
  • 157.Chablani J, Rani PK, Balakrishnan D, Jalali S. Unusual Adverse Choroidal Reaction to Intravitreal Bevacizumab in Aggressive Posterior Retinopathy of Prematurity: The Indian Twin Cities ROP Screening (ITCROPS) Data Base Report Number 7. Semin Ophthalmol. 2014;29:222–225. doi: 10.3109/08820538.2013.835842. [DOI] [PubMed] [Google Scholar]
  • 158.Tahija SG, Hersetyati R, Lam GC, Kusaka S, McMenamin PG. Fluorescein angiographic observations of peripheral retinal vessel growth in infants after intravitreal injection of bevacizumab as sole therapy for zone I and posterior zone II retinopathy of prematurity. Br J Ophthalmol. 2014;98:507–512. doi: 10.1136/bjophthalmol-2013-304109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Lepore D, Quinn GE, Molle F, et al. Intravitreal Bevacizumab versus laser treatment in type I retinopathy of prematurity. Report on fluorescein angiographic findings. Ophthalmology. 2014;121:2212–2219. doi: 10.1016/j.ophtha.2014.05.015. [DOI] [PubMed] [Google Scholar]
  • 160.Lee BJ, Kim JH, Heo H, Yu YS. Delayed onset atypical vitreoretinal traction band formation after an intravitreal injection of bevacizumab in stage 3 retinopathy of prematurity. Eye (Lond) 2012;26:903–909. doi: 10.1038/eye.2012.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Menke MN, Framme C, Nelle M, Berger MR, Sturm V, Wolf S. Intravitreal ranibizumab monotherapy to treat retinopathy of prematurity zone II, stage 3 with plus disease. BMC Ophthalmol. 2015;15:20. doi: 10.1186/s12886-015-0001-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Minami T, Kuniyoshi K, Kusaka S, et al. Intravitreal injection of bevacizumab for retinopathy of prematurity in an infant with peters anomaly. Case Rep Ophthalmol. 2014;5:318–324. doi: 10.1159/000368298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Yaz Y, Erol N, Gursoy H, Basmak H, Bilgec MD. A rare association of intravitreal bevacizumab injection with double ridge formation in retinopathy of prematurity. J Pediatr Ophthalmol Strabismus. 2014;51 doi: 10.3928/01913913-20140930-01. [DOI] [PubMed] [Google Scholar]
  • 164.Chen W, Binenbaum G, Karp K, et al. Late recurrence of retinopathy of prematurity after treatment with both intravitreal bevacizumab and laser. J AAPOS. 2014;18:402–404. doi: 10.1016/j.jaapos.2014.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Chen YH, Chen SN, Lien RI, et al. Refractive errors after the use of bevacizumab for the treatment of retinopathy of prematurity: 2-year outcomes. Eye (Lond) 2014;28:1080–1086. doi: 10.1038/eye.2014.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Mehta S, Hubbard GB., 3rd Delayed recurrent neovascularization and persistent avascular retina following intravitreal bevacizumab for retinopathy of prematurity. Retin Cases Brief Rep. 2013;7:206–209. doi: 10.1097/ICB.0b013e318285238e. [DOI] [PubMed] [Google Scholar]
  • 167.Patel RD, Blair MP, Shapiro MJ, Lichtenstein SJ. Significant treatment failure with intravitreous bevacizumab for retinopathy of prematurity. Arch Ophthalmol. 2012;130:801–802. doi: 10.1001/archophthalmol.2011.1802. [DOI] [PubMed] [Google Scholar]
  • 168.McCloskey M, Wang H, Jiang Y, Smith GW, Strange J, Hartnett ME. Anti-VEGF antibody leads to later atypical intravitreous neovascularization and activation of angiogenic pathways in a rat model of retinopathy of prematurity. Invest Ophthalmol Vis Sci. 2012;54:2020–2026. doi: 10.1167/iovs.13-11625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Sato T, Wada K, Arahori H, et al. Serum concentrations of bevacizumab (avastin) and vascular endothelial growth factor in infants with retinopathy of prematurity. Am J Ophthalmol. 2012;153:327–333. doi: 10.1016/j.ajo.2011.07.005. [DOI] [PubMed] [Google Scholar]
  • 170.Hong YR, Kim YH, Kim SY, Nam GY, Cheon HJ, Lee SJ. Plasma concentrations of vascular endothelial growth factor in retinopathy of prematurity after intravitreal bevacizumab injection. Retina. 2015;35:1772–1777. doi: 10.1097/IAE.0000000000000535. [DOI] [PubMed] [Google Scholar]
  • 171.Wu WC, Lien R, Liao PJ, et al. Serum levels of vascular endothelial growth factor and related factors after intravitreous bevacizumab injection for retinopathy of prematurity. JAMA Ophthalmol. 2015;133:391–397. doi: 10.1001/jamaophthalmol.2014.5373. [DOI] [PubMed] [Google Scholar]
  • 172.Kong L, Bhatt AR, Demny AB, et al. Pharmacokinetics of bevacizumab and its effects on serum VEGF and IGF-1 in infants with retinopathy of prematurity. Invest Ophthalmol Vis Sci. 2015;56:956–961. doi: 10.1167/iovs.14-15842. [DOI] [PubMed] [Google Scholar]
  • 173.Pieh C1, Agostini H, Buschbeck C, et al. VEGF-A, VEGFR-1, VEGFR-2 and Tie2 levels in plasma of premature infants: relationship to retinopathy of prematurity. Br J Ophthalmol. 2008;92:689–693. doi: 10.1136/bjo.2007.128371. [DOI] [PubMed] [Google Scholar]
  • 174.Hapsari D1, Sitorus RS. Intravitreal Bevacizumab in Retinopathy of Prematurity: Inject or Not? Asia Pac J Ophthalmol (Phila) 2014;3:368–378. doi: 10.1097/APO.0000000000000039. [DOI] [PubMed] [Google Scholar]
  • 175.Wong RK1, Hubschman S, Tsui I. Reactivation of retinopathy of prematurity after ranibizumab treatment. Retina. 2015;35:675–680. doi: 10.1097/IAE.0000000000000578. [DOI] [PubMed] [Google Scholar]
  • 176.Chen SN, Lian I, Hwang YC, et al. Intravitreal anti-vascular endothelial growth factor treatment for retinopathy of prematurity: comparison between ranibizumab and bevacizumab. Retina. 2015;35:667–674. doi: 10.1097/IAE.0000000000000380. [DOI] [PubMed] [Google Scholar]
  • 177.Autrata R, Senková K, Holousová M, Krejcírová I, Dolezel Z, Borek I. Effects of intravitreal pegaptanib or bevacizumab and laser in treatment of threshold retinopathy of prematurity in zone I and posterior zone II--four years results. Cesk Slov Oftalmol. 2012;68:29–36. [PubMed] [Google Scholar]
  • 178.Mintz-Hittner HA. Intravitreal pegaptanib as adjunctive treatment for stage 3+ ROP shown to be effective in a prospective, randomized, controlled multicenter clinical trial. Eur J Ophthalmol. 2012;22:685–686. doi: 10.5301/ejo.5000176. [DOI] [PubMed] [Google Scholar]
  • 179.Avery RL. Bevacizumab (Avastin) for retinopathy of prematurity: wrong dose, wrong drug, or both? J AAPOS. 2012;16:2–4. doi: 10.1016/j.jaapos.2011.11.002. [DOI] [PubMed] [Google Scholar]
  • 180.Mireskandari K, Adams GG, Tehrani NN. Recurrence of retinopathy of prematurity following bevacizumab monotherapy: is it only the tip of the iceberg? JAMA Ophthalmol. 2013;131:544–545. doi: 10.1001/jamaophthalmol.2013.711. [DOI] [PubMed] [Google Scholar]
  • 181.Darlow BA, Ells AL, Gilbert CE, Gole GA, Quinn GE. Are we there yet? Bevacizumab therapy for retinopathy of prematurity. Arch Dis Child Fetal Neonatal Ed. 2013;98:F170–F174. doi: 10.1136/archdischild-2011-301148. [DOI] [PubMed] [Google Scholar]
  • 182.Hartnett ME. Vascular endothelial growth factor antagonist therapy for retinopathy of prematurity. Clin Perinatol. 2014;41:925–943. doi: 10.1016/j.clp.2014.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Fleck BW. Management of retinopathy of prematurity. Arch Dis Child Fetal Neonatal Ed. 2013;98:F454–F456. doi: 10.1136/archdischild-2013-303933. [DOI] [PubMed] [Google Scholar]
  • 184.Mititelu M, Chaudhary KM, Lieberman RM. An evidence-based meta-analysis of vascular endothelial growth factor inhibition in pediatric retinal diseases: part 1. Retinopathy of prematurity. J Pediatr Ophthalmol Strabismus. 2012;49:332–340. doi: 10.3928/01913913-20120821-03. [DOI] [PubMed] [Google Scholar]
  • 185.Hård AL, Hellström A. On safety, pharmacokinetics and dosage of bevacizumab in ROP treatment - a review. Acta Paediatr. 2011;100:1523–1527. doi: 10.1111/j.1651-2227.2011.02445.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Hård AL, Hellström A. On the use of antiangiogenetic medications for retinopathy of prematurity. Acta Paediatr. 2011;100:1063–1065. doi: 10.1111/j.1651-2227.2011.02330.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.King GL, Goodman AD, Buzney S, Moses A, Kahn CR. Receptors and growth-promoting effects of insulin and insulin-like growth factors on cells from bovine retinal capillaries and aorta. J Clin Invest. 1985;75:1028–1036. doi: 10.1172/JCI111764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Hellstrom A, Perruzzi C, Ju M, Engstrom E, et al. Low IGF-1 suppresses VEGF-survival signaling in retinal endothelial cells: direct correlation with clinical retinopathy of prematurity. Proc Natl Acad Sci USA. 2001;98:5804–5808. doi: 10.1073/pnas.101113998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Smith LE, Shen W, Perruzzi C, et al. Regulation of vascular endothelial growth factor-dependent retinal neovascularization by insulin-like growth factor-1 receptor. Nat Med. 1999;5:1390–1395. doi: 10.1038/70963. [DOI] [PubMed] [Google Scholar]
  • 190.Smith LE. IGF-1 and retinopathy of prematurity in the preterm infant. Biol Neonate. 2005;88:237–244. doi: 10.1159/000087587. [DOI] [PubMed] [Google Scholar]
  • 191.Hellstrom A, Carlsson B, Niklasson A, et al. IGF-1 is critical for normal vascularization of the human retina. J Clin Endocrinol Metab. 2002;87:3413–3416. doi: 10.1210/jcem.87.7.8629. [DOI] [PubMed] [Google Scholar]
  • 192.Shaw LC, Grant MB. Insulin like growth factor-1 and insulin-like growth factor binding proteins: their possible roles in both maintaining normal retinal vascular function and in promoting retinal pathology. Rev Endocr Metab Disord. 2004;5:199–207. doi: 10.1023/B:REMD.0000032408.18015.b1. [DOI] [PubMed] [Google Scholar]
  • 193.Smith LE. Pathogenesis of retinopathy of prematurity. Growth Horm IGF Res. 2004;14(Suppl A):S140–S144. doi: 10.1016/j.ghir.2004.03.030. [DOI] [PubMed] [Google Scholar]
  • 194.Hellström A, Engström E, Hård AL, et al. Postnatal serum insulin-like growth factor I deficiency is associated with retinopathy of prematurity and other complications of premature birth. Pediatrics. 2003;112:1016–1020. doi: 10.1542/peds.112.5.1016. [DOI] [PubMed] [Google Scholar]
  • 195.Pérez-Muñuzuri A, Fernández-Lorenzo JR, Couce-Pico ML, Blanco-Teijeiro MJ, Fraga-Bermúdez JM. Serum levels of IGF1 are a useful predictor of retinopathy of prematurity. Acta Paediatr. 2010;99:519–525. doi: 10.1111/j.1651-2227.2009.01677.x. [DOI] [PubMed] [Google Scholar]
  • 196.Can E, Bülbül A, Uslu S, Bolat F, Cömert S, Nuhoğlu A. Early Aggressive parenteral nutrition induced high insulin-like growth factor 1 (IGF-1) and insulin-like growth factor binding protein 3 (IGFBP3) levels can prevent risk of retinopathy of prematurity. Iran J Pediatr. 2013;23:403–410. [PMC free article] [PubMed] [Google Scholar]
  • 197.Kant S, Usha, Bhatia, Anthony K, Seth G. Pattern of retinopathy of prematurity and its correlation with insulin-like growth factor-1 (IGF-1) Ann Ophthalmol (Skokie) 2009;41:157–161. [PubMed] [Google Scholar]
  • 198.Reddy MA, Patel HI, Karim SM, et al. Reduced utility of serum IGF-1 levels in predicting retinopathy of prematurity reflects maternal ethnicity. Br J Ophthalmol. 2015 doi: 10.1136/bjophthalmol-2015-307234. Ahead of Print. [DOI] [PubMed] [Google Scholar]
  • 199.Peirovifar A, Gharehbaghi MM, Gharabaghi PM, Sadeghi K. Vascular endothelial growth factor and insulin-like growth factor-1 in preterm infants with retinopathy of prematurity. Singapore Med J. 2013;54:709–712. doi: 10.11622/smedj.2013246. [DOI] [PubMed] [Google Scholar]
  • 200.Ley D, Hansen-Pupp I, Niklasson A, et al. Longitudinal infusion of a complex of insulin-like growth factor-I and IGF-binding protein-3 in five preterm infants: pharmacokinetics and short-term safety. Pediatr Res. 2013;73:68–74. doi: 10.1038/pr.2012.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Azrad M, Turgeon C, Demark-Wahnefried W. Current evidence linking polyunsaturated Fatty acids with cancer risk and progression. Front Oncol. 2013;3:224. doi: 10.3389/fonc.2013.00224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Yazdi PG. A review of the biologic and pharmacologic role of docosapentaenoic acid n-3. Version 2. F1000Res. 2013;2:256. doi: 10.12688/f1000research.2-256.v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Harris WS, Baack ML. Beyond building better brains: bridging the docosahexaenoic acid (DHA) gap of prematurity. J Perinatol. 2015;35:1–7. doi: 10.1038/jp.2014.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Crawford MA, Costeloe K, Ghebremeskel K, Phylactos A, Skirvin L, Stacey F. Are deficits of arachidonic and docosahexaenoic acids responsible for the neural and vascular complications of preterm babies? Am J Clin Nutr. 1997;66:1032S–1041S. doi: 10.1093/ajcn/66.4.1032S. [DOI] [PubMed] [Google Scholar]
  • 205.Smith WL. Nutritionally essential fatty acids and biologically indispensable cyclooxygenases. Trends Biochem Sci. 2008;33:27–37. doi: 10.1016/j.tibs.2007.09.013. [DOI] [PubMed] [Google Scholar]
  • 206.Wang W, Zhu J, Lyu F, et al. ω-3 polyunsaturated fatty acids-derived lipid metabolites on angiogenesis, inflammation and cancer. Prostaglandins Other Lipid Mediat. 2014;113–115:13–20. doi: 10.1016/j.prostaglandins.2014.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Yang P, Jiang Y, Fischer SM. Prostaglandin E3 metabolism and cancer. Cancer Lett. 2014;348:1–11. doi: 10.1016/j.canlet.2014.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Arnold C, Konkel A, Fischer R, Schunck WH. Cytochrome P450-dependent metabolism of omega-6 and omega-3 long-chain polyunsaturated fatty acids. Pharmacol Rep. 2010;62:536–547. doi: 10.1016/s1734-1140(10)70311-x. [DOI] [PubMed] [Google Scholar]
  • 209.Smith WL. Cyclooxygenases, peroxide tone and the allure of fish oil. Curr Opin Cell Biol. 2005;17:174–182. doi: 10.1016/j.ceb.2005.02.005. [DOI] [PubMed] [Google Scholar]
  • 210.Rombaldi Bernardi J, de Souza Escobar R, Ferreira CF, Pelufo Silveira P. Fetal and neonatal levels of omega-3: effects on neurodevelopment, nutrition, and growth. Scientific World Journal. 2012;2012:202473. doi: 10.1100/2012/202473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Abeywardena MY, Head RJ. Long chain n-3 polyunsaturated fatty acids and blood vessel function. Cardiovasc Res. 2001;52:361–371. doi: 10.1016/s0008-6363(01)00406-0. [DOI] [PubMed] [Google Scholar]
  • 212.SanGiovanni JP, Chew EY. The role of omega-3 long-chain polyunsaturated fatty acids in health and disease of the retina. Prog Retin Eye Res. 2005;24:87–138. doi: 10.1016/j.preteyeres.2004.06.002. [DOI] [PubMed] [Google Scholar]
  • 213.Pawlik D, Lauterbach R, Walczak M, Hurkała J, Sherman MP. Fish-oil fat emulsion supplementation reduces the risk of retinopathy in very low birth weight infants: a prospective, randomized study. JPEN J Parenter Enteral Nutr. 2014;38:711–716. doi: 10.1177/0148607113499373. [DOI] [PubMed] [Google Scholar]
  • 214.Pawlik D, Lauterbach R, Turyk E. Fish-oil fat emulsion supplementation may reduce the risk of severe retinopathy in VLBW infants. Pediatrics. 2011;127:223–228. doi: 10.1542/peds.2010-2427. [DOI] [PubMed] [Google Scholar]
  • 215.Beken S1, Dilli D2, Fettah ND2, Kabataş EU3, Zenciroğlu A2, Okumuş N2. The influence of fish-oil lipid emulsions on retinopathy of prematurity in very low birth weight infants: a randomized controlled trial. Early Hum Dev. 2014 Jan;90(1):27–31. doi: 10.1016/j.earlhumdev.2013.11.002. [DOI] [PubMed] [Google Scholar]
  • 216.Simmer K, Rao SC. Early introduction of lipids to parenterally-fed preterm infants. Cochrane Database Syst Rev. 2005;(2):CD005256. doi: 10.1002/14651858.CD005256. [DOI] [PubMed] [Google Scholar]
  • 217.Smithers LG, Gibson RA, McPhee A, Makrides M. Effect of long-chain polyunsaturated fatty acid supplementation of preterm infants on disease risk and neurodevelopment: a systematic review of randomized controlled trials. Am J Clin Nutr. 2008;87:912–920. doi: 10.1093/ajcn/87.4.912. [DOI] [PubMed] [Google Scholar]
  • 218.Holmes JM, Cronin CM, Squires P, Myers TF. Randomized clinical trial of surfactant prophylaxis in retinopathy of prematurity. J Pediatr Ophthalmol Strabismus. 1994;31:189–191. doi: 10.3928/0191-3913-19940501-11. [DOI] [PubMed] [Google Scholar]
  • 219.Termote JU, Schalij-Delfos NE, Wittebol-Post D, Brouwers HA, Hoogervorst BR, Cats BP. Surfactant replacement therapy: a new risk factor in developing retinopathy of prematurity? Eur J Pediatr. 1994;153:113–116. doi: 10.1007/BF01959221. [DOI] [PubMed] [Google Scholar]
  • 220.Modanlou HD1, Beharry K, Padilla G, Norris K, Safvati S, Aranda JV. Comparative efficacy of Exosurf and Survanta surfactants on early clinical course of respiratory distress syndrome and complications of prematurity. J Perinatol. 1997;17:455–460. [PubMed] [Google Scholar]
  • 221.Halliday HL. Overview of clinical trials comparing natural and synthetic surfactants. Biol Neonate. 1995;67(Suppl 1):32–47. doi: 10.1159/000244205. [DOI] [PubMed] [Google Scholar]
  • 222.Romagnoli C, Tesfagabir MG, Giannantonio C, Papacci P. Erythropoietin and retinopathy of prematurity. Early Hum Dev. 2011;87(Suppl 1):S39–S42. doi: 10.1016/j.earlhumdev.2011.01.027. [DOI] [PubMed] [Google Scholar]
  • 223.Brown MS, Barón AE, France EK, Hamman RF. Association between higher cumulative doses of recombinant erythropoietin and risk for retinopathy of prematurity. J AAPOS. 2006;10:143–149. doi: 10.1016/j.jaapos.2005.09.005. [DOI] [PubMed] [Google Scholar]
  • 224.Suk KK, Dunbar JA, Liu A, et al. Human recombinant erythropoietin and the incidence of retinopathy of prematurity: a multiple regression model. J AAPOS. 2008;12:233–238. doi: 10.1016/j.jaapos.2007.08.009. [DOI] [PubMed] [Google Scholar]
  • 225.Sato T, Kusaka S, Shimojo H, Fujikado T. Vitreous levels of erythropoietin and vascular endothelial growth factor in eyes with retinopathy of prematurity. Ophthalmology. 2009;116:1599–1603. doi: 10.1016/j.ophtha.2008.12.023. [DOI] [PubMed] [Google Scholar]
  • 226.Figueras-Aloy J, Alvarez-Domínguez E, Morales-Ballus M, Salvia-Roiges MD, Moretones-Suñol G. Early administration of erythropoietin in the extreme premature, a risk factor for retinopathy of prematurity? An Pediatr (Barc) 2010;73:327–333. doi: 10.1016/j.anpedi.2010.09.001. [DOI] [PubMed] [Google Scholar]
  • 227.Kandasamy Y, Kumar P, Hartley L. The effect of erythropoietin on the severity of retinopathy of prematurity. Eye (Lond) 2014;28:814–818. doi: 10.1038/eye.2014.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Manzoni P, Memo L, Mostert M, et al. Use of erythropoietin is associated with threshold retinopathy of prematurity (ROP) in preterm ELBW neonates: a retrospective, cohort study from two large tertiary NICUs in Italy. Early Hum Dev. 2014;90(Suppl 2):S29–S33. doi: 10.1016/S0378-3782(14)50009-6. [DOI] [PubMed] [Google Scholar]
  • 229.Schneider JK, Gardner DK, Cordero L. Use of recombinant human erythropoietin and risk of severe retinopathy in extremely low-birth-weight infants. Pharmacotherapy. 2008;28:1335–1340. doi: 10.1592/phco.28.11.1335. [DOI] [PubMed] [Google Scholar]
  • 230.Shah N, Jadav P, Jean-Baptiste D, Weedon J, Cohen LM, Kim MR. The effect of recombinant human erythropoietin on the development of retinopathy of prematurity. Am J Perinatol. 2010;27:67–71. doi: 10.1055/s-0029-1224872. [DOI] [PubMed] [Google Scholar]
  • 231.Fauchère JC, Koller BM, Tschopp A, Dame C, Ruegger C, Bucher HU Swiss Erythropoietin Neuroprotection Trial Group. Safety of early high-dose recombinant erythropoietin for neuroprotection in very preterm infants. J Pediatr. 2015;167:52–57. doi: 10.1016/j.jpeds.2015.02.052. [DOI] [PubMed] [Google Scholar]
  • 232.Ohlsson A, Aher SM. Early erythropoietin for preventing red blood cell transfusion in preterm and/or low birth weight infants. Cochrane Database Syst Rev. 2006;(3):CD004863. doi: 10.1002/14651858.CD004863.pub2. [DOI] [PubMed] [Google Scholar]
  • 233.Ohlsson A, Aher SM. Early erythropoietin for preventing red blood cell transfusion in preterm and/or low birth weight infants. Cochrane Database Syst Rev. 2014;4:CD004863. doi: 10.1002/14651858.CD004863.pub4. [DOI] [PubMed] [Google Scholar]
  • 234.Wang H, Zhang L, Jin Y. A meta-analysis of the protective effect of recombinant human erythropoietin (rhEPO) for neurodevelopment in preterm infants. A meta-analysis of the protective effect of recombinant human erythropoietin (rhEPO) for neurodevelopment in preterm infants. Cell Biochem Biophys. 2015;71:795–802. doi: 10.1007/s12013-014-0265-1. [DOI] [PubMed] [Google Scholar]
  • 235.Gess B, Sandner P, Kurtz A. Differential effects of kinase inhibitors on erythropoietin and vascular endothelial growth factor gene expression in rat hepatocytes. Pflugers Arch. 1996;432:426–432. doi: 10.1007/s004240050154. [DOI] [PubMed] [Google Scholar]
  • 236.Krantz SB. Erythropoietin. Blood. 1991;77:419–434. [PubMed] [Google Scholar]
  • 237.Batton DG, Roberts C, Trese M, Maisels MJ. Severe retinopathy of prematurity and steroid exposure. Pediatrics. 1992;90:534–536. [PubMed] [Google Scholar]
  • 238.Sobel DB, Philip AG. Prolonged dexamethasone therapy reduces the incidence of cryotherapy for retinopathy of prematurity in infants of less than 1 kilogram birth weight with bronchopulmonary dysplasia. Pediatrics. 1992;90:529–533. [PubMed] [Google Scholar]
  • 239.Todd DA, Kennedy J, Roberts S, Watts J, Psaila K. John Retinopathy of prematurity in infants less than 29 weeks' gestation at birth. Aust N Z J Ophthalmol. 1994;22:19–23. doi: 10.1111/j.1442-9071.1994.tb01690.x. [DOI] [PubMed] [Google Scholar]
  • 240.Ramanathan R, Siassi B, deLemos RA. Severe retinopathy of prematurity in extremely low birth weight infants after short-term dexamethasone therapy. J Perinatol. 1995;15:178–182. [PubMed] [Google Scholar]
  • 241.Haroon Parupia MF, Dhanireddy R. Association of postnatal dexamethasone use and fungal sepsis in the development of severe retinopathy of prematurity and progression to laser therapy in extremely low-birth-weight infants. J Perinatol. 2001;21:242–247. doi: 10.1038/sj.jp.7200531. [DOI] [PubMed] [Google Scholar]
  • 242.Smolkin T, Steinberg M, Sujov P, Mezer E, Tamir A, Makhoul IR. Late postnatal systemic steroids predispose to retinopathy of prematurity in very-low-birth-weight infants: a comparative study. Acta Paediatr. 2008;97:322–326. doi: 10.1111/j.1651-2227.2008.00629.x. [DOI] [PubMed] [Google Scholar]
  • 243.Karna P, Muttineni J, Angell L, Karmaus W. Retinopathy of prematurity and risk factors: a prospective cohort study. BMC Pediatr. 2005;5:18. doi: 10.1186/1471-2431-5-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Hoogerwerf A, Schalij-Delfos NE, van Schooneveld MJ, Termote JU. Incidence of retinopathy of prematurity over the last decade in the Central Netherlands. Neonatology. 2010;98:137–142. doi: 10.1159/000280386. [DOI] [PubMed] [Google Scholar]
  • 245.Wright K, Wright SP. Lack of association of glucocorticoid therapy and retinopathy of prematurity. Arch Pediatr Adolesc Med. 1994;148:848–852. doi: 10.1001/archpedi.1994.02170080078015. [DOI] [PubMed] [Google Scholar]
  • 246.Cuculich PS, DeLozier KA, Mellen BG, Shenai JP. Postnatal dexamethasone treatment and retinopathy of prematurity in very-low-birth-weight neonates. Biol Neonate. 2001;79:9–14. doi: 10.1159/000047059. [DOI] [PubMed] [Google Scholar]
  • 247.Kothadia JM, O'Shea TM, Roberts D, Auringer ST, Weaver RG, 3rd, Dillard RG. Randomized placebo-controlled trial of a 42-Day tapering course of dexamethasone to reduce the duration of ventilator dependency in very low birth weight infants. Pediatrics. 1999;104:22–27. doi: 10.1542/peds.104.1.22. [DOI] [PubMed] [Google Scholar]
  • 248.Jacobs MA, Blanco CE. The incidence of retinopathy of prematurity and steroid therapy for broncho-pulmonary dysplasia. J Perinat Med. 1998;26:13–16. doi: 10.1515/jpme.1998.26.1.13. [DOI] [PubMed] [Google Scholar]
  • 249.Durand M, Sardesai S, McEvoy C. Effects of early dexamethasone therapy on pulmonary mechanics and chronic lung disease in very low birth weight infants: a randomized, controlled trial. Pediatrics. 1995;95:584–590. [PubMed] [Google Scholar]
  • 250.Halliday HL, Ehrenkranz RA. Early postnatal (<96 hours) corticosteroids for preventing chronic lung disease in preterm infants. Cochrane Database Syst Rev. 2000;(2):CD001146. doi: 10.1002/14651858.CD001146. [DOI] [PubMed] [Google Scholar]
  • 251.Halliday HL1, Ehrenkranz RA, Doyle LW. Early (< 8 days) postnatal corticosteroids for preventing chronic lung disease in preterm infants. Cochrane Database Syst Rev. 2010;(1):CD001146. doi: 10.1002/14651858.CD001146.pub3. [DOI] [PubMed] [Google Scholar]
  • 252.Doyle LW, Ehrenkranz RA, Halliday HL. Late (>7 days) postnatal corticosteroids for chronic lung disease in preterm infants. Cochrane Database Syst Rev. 2014;5:CD001145. doi: 10.1002/14651858.CD001145.pub3. [DOI] [PubMed] [Google Scholar]
  • 253.Halliday HL, Ehrenkranz RA. Delayed (>3 weeks) postnatal corticosteroids for chronic lung disease in preterm infants. Cochrane Database Syst Rev. 2000;(2):CD001145. doi: 10.1002/14651858.CD001145. [DOI] [PubMed] [Google Scholar]
  • 254.Higgins RD, Mendelsohn AL, DeFeo MJ, Ucsel R, Hendricks-Munoz KD. Antenatal dexamethasone and decreased severity of retinopathy of prematurity. Arch Ophthalmol. 1998;116:601–605. doi: 10.1001/archopht.116.5.601. [DOI] [PubMed] [Google Scholar]
  • 255.Vento M, Aguar M, Escobar J, et al. Antenatal steroids and antioxidant enzyme activity in preterm infants: influence of gender and timing. Antioxid Redox Signal. 2009;11:2945–2955. doi: 10.1089/ars.2009.2671. [DOI] [PubMed] [Google Scholar]
  • 256.Lee BH, Stoll BJ, McDonald SA, Higgins RD National Institute of Child Health and Human Development Neonatal Research Network. Adverse neonatal outcomes associated with antenatal dexamethasone versus antenatal betamethasone. Pediatrics. 2006;117:1503–1510. doi: 10.1542/peds.2005-1749. [DOI] [PubMed] [Google Scholar]
  • 257.Procianoy RS, Garcia-Prats JA, Hittner HM, Adams JM, Rudolph AJ. Use of indomethacin and its relationship to retinopathy of prematurity in very low birthweight infants. Arch Dis Child. 1980;55:362–364. doi: 10.1136/adc.55.5.362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Yeh TF, Raval D, Pyati S, Pildes RS. Retinopathy of prematurity (ROP) and indomethacin therapy in premature infants with patent ductus arteriosus (PDA) Prostaglandins. 1983;25:385–391. doi: 10.1016/0090-6980(83)90041-2. [DOI] [PubMed] [Google Scholar]
  • 259.Laughon M, Bose C, Clark R. Treatment strategies to prevent or close a patent ductus arteriosus in preterm infants and outcomes. J Perinatol. 2007;27:164–170. doi: 10.1038/sj.jp.7211662. [DOI] [PubMed] [Google Scholar]
  • 260.Darlow BA, Horwood LJ, Clemett RS. Retinopathy of prematurity: risk factors in a prospective population-based study. Paediatr Perinat Epidemiol. 1992;6:62–80. doi: 10.1111/j.1365-3016.1992.tb00747.x. [DOI] [PubMed] [Google Scholar]
  • 261.Jegatheesan P, Ianus V, Buchh B, et al. Increased indomethacin dosing for persistent patent ductus arteriosus in preterm infants: a multicenter, randomized, controlled trial. J Pediatr. 2008;153:183–189. doi: 10.1016/j.jpeds.2008.01.031. [DOI] [PubMed] [Google Scholar]
  • 262.Goldman RD, Spierer A, Zhurkovsky A, Kwint J, Schwarcz M, Ben Simon GJ. Retinopathy of prematurity in very low birth weight infants and the potential protective role of indomethacin. Ophthalmic Surg Lasers Imaging. 2010;41:41–47. doi: 10.3928/15428877-20091230-08. [DOI] [PubMed] [Google Scholar]
  • 263.Hammerman C. Indomethacin and retinopathy of prematurity: the hidden paradox. J Pediatr. 2008;153:587–588. doi: 10.1016/j.jpeds.2008.05.009. [DOI] [PubMed] [Google Scholar]

RESOURCES