rhIGF-1/rhIGFBP-3 in Preterm Infants: A Phase 2 Randomized Controlled Trial

David Ley, Boubou Hallberg, Ingrid Hansen-Pupp, Carlo Dani, Luca A Ramenghi, Neil Marlow, Kathryn Beardsall, Faizah Bhatti, David Dunger, Jason D Higginson, Ajit Mahaveer, Olachi J Mezu-Ndubuisi, Peter Reynolds, Carmen Giannantonio, Mirjam van Weissenbruch, Norman Barton, Adina Tocoian, Mohamed Hamdani, Emily Jochim, Alexandra Mangili, Jou-Ku Chung, Mark A Turner, Lois E H Smith, Ann Hellström, study team, David Ley, Boubou Hallberg, Ingrid Hansen-Pupp, Carlo Dani, Luca A Ramenghi, Neil Marlow, Kathryn Beardsall, Faizah Bhatti, David Dunger, Jason D Higginson, Ajit Mahaveer, Olachi J Mezu-Ndubuisi, Peter Reynolds, Carmen Giannantonio, Mirjam van Weissenbruch, Norman Barton, Adina Tocoian, Mohamed Hamdani, Emily Jochim, Alexandra Mangili, Jou-Ku Chung, Mark A Turner, Lois E H Smith, Ann Hellström, study team

Abstract

Objective: To investigate recombinant human insulin-like growth factor 1 complexed with its binding protein (rhIGF-1/rhIGFBP-3) for the prevention of retinopathy of prematurity (ROP) and other complications of prematurity among extremely preterm infants.

Study design: This phase 2 trial was conducted from September 2014 to March 2016. Infants born at a gestational age of 230/7 weeks to 276/7 weeks were randomly allocated to rhIGF-1/rhIGFBP-3 (250 µg/kg/ 24 hours, continuous intravenous infusion from <24 hours of birth to postmenstrual age 296/7 weeks) or standard neonatal care, with follow-up to a postmenstrual age of 404/7 weeks. Target exposure was ≥70% IGF-1 measurements within 28-109 µg/L and ≥70% intended therapy duration. The primary endpoint was maximum severity of ROP. Secondary endpoints included time to discharge from neonatal care, bronchopulmonary dysplasia, intraventricular hemorrhage, and growth measures.

Results: Overall, 61 infants were allocated to rhIGF-1/rhIGFBP-3, 60 to standard care (full analysis set); 24 of 61 treated infants achieved target exposure (evaluable set). rhIGF-1/rhIGFBP-3 did not decrease ROP severity or ROP occurrence. There was, however, a 53% decrease in severe bronchopulmonary dysplasia in the full analysis set (21.3% treated vs 44.9% standard care), and an 89% decrease in the evaluable set (4.8% vs 44.9%; P = .04 and P = .02, respectively) for severity distribution between groups. There was also a nonsignificant trend toward decrease in grades 3-4 intraventricular hemorrhage in the full analysis set (13.1% vs 23.3%) and in the evaluable set (8.3% vs 23.3%). Fatal serious adverse events were reported in 19.7% of treated infants (12/61) and 11.7% of control infants (7/60). No effect was observed on time to discharge from neonatal care/growth measures.

Conclusions: rhIGF-1/rhIGFBP-3 did not affect development of ROP, but decreased the occurrence of severe bronchopulmonary dysplasia, with a nonsignificant decrease in grades 3-4 intraventricular hemorrhage.

Trial registration: ClinicalTrials.gov: NCT01096784.

Keywords: bronchopulmonary dysplasia; intraventricular hemorrhage; neonatology; retinopathy of prematurity.

Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.
Figure 1.
A, Study design and B, patient disposition. *Informed consent was obtained before birth or within 24 hours after birth. † One infant had an SAE with a fatal outcome, but the primary reason for discontinuation was withdrawal of consent. ‡ All infants discontinued owing to an SAE with fatal outcome. § Seven of 9 discontinuations were owing to SAEs with fatal outcome.
Figure 2.
Figure 2.
Mean (SD) serum IGF-1 concentrations over time in infants in the standard neonatal care and rhIGF-1/rhIGFBP-3 groups (n = 121).
Figure 3.
Figure 3.
IGF-1 levels by A, ROP severity* and B, BPD severity† by postnatal week. *Mean (±SE) serum IGF-1 levels and ROP severity (<3, ≥3) in the rhIGF-1/rhIGFBP-3 and standard neonatal care groups by postnatal week. †Mean (±SE) serum IGF-1 levels and BPD severity (mild, moderate, or severe) in the rhIGF-1/rhIGFBP-3 and standard neonatal care groups by postnatal week. Note: If an infant had multiple IGF-1 levels in a day, then IGF-1 level was averaged for the day.
Figure 4.
Figure 4.
A, Average weight, B, length, and C, head circumference by treatment group (FAS).

References

    1. Langford K, Nicolaides K, Miell JP. Maternal and fetal insulin-like growth factors and their binding proteins in the second and third trimesters of human pregnancy. Hum Reprod 1998;13:1389–93.
    1. Hellström A, Ley D, Hansen-Pupp I, Hallberg B, Löfqvist C, van Marter L, et al. Insulin-like growth factor 1 has multisystem effects on foetal and preterm infant development. Acta Paediatr 2016;105:576–86.
    1. Hansen-Pupp I, Löfqvist C, Polberger S, Niklasson A, Fellman V, Hellstrom A, et al. Influence of insulin-like growth factor I and nutrition during phases of postnatal growth in very preterm infants. Pediatr Res 2011;69:448–53.
    1. Lineham JD, Smith RM, Dahlenburg GW, King RA, Haslam RR, Stuart MC, et al. Circulating insulin-like growth factor I levels in newborn premature and full-term infants followed longitudinally. Early Hum Dev 1986;13:37–46.
    1. Hellström A, Engstrom E, Hård AL, Albertsson-Wikland K, Carlsson B, Niklasson A, et al. Postnatal serum insulin-like growth factor I deficiency is associated with retinopathy of prematurity and other complications of premature birth. Pediatrics 2003; 112:1016–20.
    1. 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;99:519–25.
    1. Löfqvist C, Engström E, Sigurdsson J, Hard AL, Niklasson A, Ewald U, et al. Postnatal head growth deficit among premature infants parallels retinopathy of prematurity and insulin-like growth factor-1 deficit. Pediatrics 2006;117:1930–8.
    1. Löfqvist C, Hellgren G, Niklasson A, Engström E, Ley D, Hansen-Pupp I. Low postnatal serum IGF-I levels are associated with bronchopulmonary dysplasia (BPD). Acta Paediatr 2012;101:1211–6.
    1. Klevebro S, Lundgren P, Hammar U, Smith LE, Bottai M, Domellof M, et al. Cohort study of growth patterns by gestational age in preterm infants developing morbidity. BMJ Open 2016;6:e012872.
    1. Hellstrom A, Perruzzi C, Ju M, Engström E, Hård AL, Liu JL, et al. Low IGF-I suppresses VEGF-survival signaling in retinal endothelial cells: direct correlation with clinical retinopathy of prematurity. Proc Natl Acad Sci USA 2001;98:5804–8.
    1. Vanhaesebrouck S, Daniëls H, Moons L, Vanhole C, Carmeliet P, De Zegher F. Oxygen-induced retinopathy in mice: amplification by neonatal IGF-I deficit and attenuation by IGF-I administration. Pediatr Res 2009;65:307–10.
    1. Jin ZA, Jin ZY, Chi YX, Lu JR. Effects of recombinant human insulin-like growth factor-1 on the expression of Clara cell secretory protein in lung of hyperoxia-exposed newborn rats. Zhonghua Er Ke Za Zhi 2007;45:369–73. [in Chinese].
    1. Jakkula M, Le Cras TD, Gebb S, Hirth KP, Tuder RM, Voelkel NF, et al. Inhibition of angiogenesis decreases alveolarization in the developing rat lung. Am J Physiol Lung Cell Mol Physiol 2000;279:L600–7.
    1. De Paepe ME, Mao Q, Powell J, Rubin SE, DeKoninck P, Appel N, et al. Growth of pulmonary microvasculature in ventilated preterm infants. Am J Respir Crit Care Med 2006;173:204–11.
    1. Lekic T, Flores J, Klebe D, Doycheva D, Rolland WB, Tang J, et al. Intra-nasal IGF-1 reduced rat pup germinal matrix hemorrhage. Acta Neurochir Suppl 2016;121:209–12.
    1. Ley D, Hansen-Pupp I, Niklasson A, Domellof M, Friberg LE, Borg J, 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.
    1. Hallberg B, Hansen-Pupp I, Chung J-K, Sharma J, Fetterly G, Mascelli M, et al. Determination of optimal dosing of recombinant human IGF-1/IGFBP-3 to establish and maintain physiological intrauterine serum IGF-1 levels in preterm infants. J Pediatr Neonat Individual Med 2015; 4: e040212.
    1. Lassarre C, Hardouin S, Daffos F, Forestier F, Frankenne F, Binoux M. Serum insulin-like growth factors and insulin-like growth factor binding proteins in the human fetus: relationships with growth in normal subjects and in subjects with intrauterine growth retardation. Pediatr Res 1991;29:219–25.
    1. Bang P, Westgren M, Schwander J, Blum WF, Rosenfeld RG, Stangenberg M. Ontogeny of insulin-like growth factor-binding protein-1, −2, and −3: quantitative measurements by radioimmunoassay in human fetal serum. Pediatr Res 1994;36:528–36.
    1. Chung J-K, Hallberg B, Hansen-Pupp I, Graham MA, Fetterly G, Sharma J, et al. Development and verification of a pharmacokinetic model to optimize physiologic replacement of rhIGF-1/rhIGFBP-3 in preterm infants. Pediatr Res 2017;81:504–10.
    1. International Committee for the Classification of Retinopathy of Prematurity. The International Classification of Retinopathy of Prematurity revisited. Arch Ophthalmol 2005;123:991–9.
    1. Early Treatment For Retinopathy Of Prematurity Cooperative Group. Revised indications for the treatment of retinopathy of prematurity: results of the early treatment for retinopathy of prematurity randomized trial. Arch Ophthalmol 2003;121:1684–94.
    1. Walsh MC, Szefler S, Davis J, Allen M, Van Marter L, Abman S, et al. Summary proceedings from the bronchopulmonary dysplasia group. Pediatrics 2006;117:S52–6.
    1. Jobe AH, Bancalari E. Bronchopulmonary dysplasia. Am J Respir Crit Care Med 2001;163:1723–9.
    1. Papile LA, Burstein J, Burstein R, Koffler H. Incidence and evolution of subependymal and intraventricular hemorrhage: a study of infants with birth weights less than 1,500 gm. J Pediatr 1978;92:529–34.
    1. Bowerman RA, Donn SM, Silver TM, Jaffe MH. Natural history of neonatal periventricular/intraventricular hemorrhage and its complications: sonographic observations. AJR Am J Roentgenol 1984;143:1041–52.
    1. Austeng D, Kallen KB, Ewald UW, Jakobsson PG, Holmstrom GE. Incidence of retinopathy of prematurity in infants born before 27 weeks’ gestation in Sweden. Arch Ophthalmol 2009;127:1315–9.
    1. Higgins RD, Jobe AH, Koso-Thomas M, Bancalari E, Viscardi RM, Hartert TV, et al. Bronchopulmonary dysplasia: executive summary of a workshop. J Pediatr 2018;197:300–8.
    1. Carlo WA, Finer NN, Walsh MC, Rich W, Gantz MG, Laptook AR, et al. Target ranges of oxygen saturation in extremely preterm infants. N Engl J Med 2010;362:1959–69.
    1. Stenson BJ, Tarnow-Mordi WO, Darlow BA, Simes J, Juszczak E, Askie L, et al. Oxygen saturation and outcomes in preterm infants. N Engl J Med 2013;368:2094–104.
    1. Schmidt B, Whyte RK, Asztalos EV, Moddemann D, Poets C, Rabi Y, et al. Effects of targeting higher vs lower arterial oxygen saturations on death or disability in extremely preterm infants: a randomized clinical trial. JAMA 2013;309:2111–20.
    1. Manley BJ, Kuschel CA, Elder JE, Doyle LW, Davis PG. Higher rates of retinopathy of prematurity after increasing oxygen saturation targets for very preterm infants: experience in a single center. J Pediatr 2016;168:242–4.
    1. Lundgren P, Hard AL, Wilde A, Lofqvist C, Smith LEH, Hellstrom A. Implementing higher oxygen saturation targets reduced the impact of poor weight gain as a predictor for retinopathy of prematurity. Acta Paediatr 2018;107:767–73.
    1. Hansen-Pupp I, Hellström-Westas L, Cilio CM, Andersson S, Fellman V, Ley D. Inflammation at birth and the insulin-like growth factor system in very preterm infants. Acta Paediatr 2007;96:830–6.
    1. Hansen-Pupp I, Hellstrom A, Hamdani M, Tocoian A, Kreher NC, Ley D, et al. Continuous longitudinal infusion of rhIGF-1/rhIGFBP-3 in extremely preterm infants: evaluation of feasibility in a phase II study. Growth Horm IGF Res 2017;36:44–51.
    1. Stoll BJ, Hansen NI, Bell EF, Shankaran S, Laptook AR, Walsh MC, et al. Neonatal outcomes of extremely preterm infants from the NICHD Neonatal Research Network. Pediatrics 2010;126:443–56.
    1. Stoll BJ, Hansen NI, Bell EF, Walsh MC, Carlo WA, Shankaran S, et al. Trends in care practices, morbidity, and mortality of extremely preterm neonates, 1993–2012. JAMA 2015;314:1039–51.
    1. Bowker RM, Yan X, Managlia E, Liu SXL, Marek C, Tan XD, et al. Dimethyloxalylglycine preserves the intestinal microvasculature and protects against intestinal injury in a neonatal mouse NEC model: role of VEGF signaling. Pediatr Res 2018;83:545–53.
    1. Dong CX, Zhao W, Solomon C, Rowland KJ, Ackerley C, Robine S, et al. The intestinal epithelial insulin-like growth factor-1 receptor links glucagon-like peptide-2 action to gut barrier function. Endocrinology 2014;155:370–9.
    1. Drucker DJ, DeForest L, Brubaker PL. Intestinal response to growth factors administered alone or in combination with human [Gly2]glucagon-like peptide 2. Am J Physiol 1997;273:G1252–62.
    1. Tian R, Liu SX, Williams C, Soltau TD, Dimmitt R, Zheng X, et al. Characterization of a necrotizing enterocolitis model in newborn mice. Int J Clin Exp Med 2010;3:293–302.
    1. Yan X, Managlia E, Liu SX, Tan XD, Wang X, Marek C, et al. Lack of VEGFR2 signaling causes maldevelopment of the intestinal microvasculature and facilitates necrotizing enterocolitis in neonatal mice. Am J Physiol Gastrointest Liver Physiol 2016;310:G716–25.
    1. Beardsall K, Vanhaesebrouck S, Frystyk J, Ogilvy-Stuart AL, Vanhole C, van Weissenbruch M, et al. Relationship between insulin-like growth factor I levels, early insulin treatment, and clinical outcomes of very low birth weight infants. J Pediatr 2014;164:1038–44, e1.
    1. Bassan H, Benson CB, Limperopoulos C, Feldman HA, Ringer SA, Veracruz E, et al. Ultrasonographic features and severity scoring of periventricular hemorrhagic infarction in relation to risk factors and outcome. Pediatrics 2006;117:2111–8.
    1. Dudink J, Lequin M, Weisglas-Kuperus N, Conneman N, van Goudoever JB, Govaert P. Venous subtypes of preterm periventricular haemorrhagic infarction. Arch Dis Child Fetal Neonatal Ed 2008;93:F201–6.
    1. Gough A, Spence D, Linden M, Halliday HL, McGarvey LP. General and respiratory health outcomes in adult survivors of bronchopulmonary dysplasia: a systematic review. Chest 2012;141:1554–67.
    1. Gough A, Linden MA, Spence D, Halliday HL, Patterson CC, McGarvey L. Executive functioning deficits in young adult survivors of broncho-pulmonary dysplasia. Disabil Rehabil 2015;37:1940–5.
    1. Iyengar A, Davis JM. Drug therapy for the prevention and treatment of bronchopulmonary dysplasia. Front Pharmacol 2015;6:12.
    1. McCrea HJ, Ment LR. The diagnosis, management, and postnatal prevention of intraventricular hemorrhage in the preterm neonate. Clin Perinatol 2008;35:777–92.
    1. Chung J-K, Hallberg B, Hansen-Pupp I, Graham MA, Fetterly G, Sharma J, et al. Development and verification of a pharmacokinetic model to optimize physiologic replacement of rhIGF-1/rhIGFBP-3 in preterm infants. Pediatr Res 2017;81:504–10.
    1. Hansen-Pupp I, Engstrom E, Niklasson A, Berg AC, Fellman V, Lofqvist C, et al. Fresh-frozen plasma as a source of exogenous insulin-like growth factor-I in the extremely preterm infant. J Clin Endocrinol Metab 2009;94:477–82.
    1. Blum WF, Breier BH. Radioimmunoassays for IGFs and IGFBPs. Growth Regul 1994;1(Suppl 1):11–9.

Source: PubMed

3
S'abonner