Hostname: page-component-5c6d5d7d68-qks25 Total loading time: 0 Render date: 2024-08-15T13:46:43.118Z Has data issue: false hasContentIssue false

Oral DHA supplementation and retinopathy of prematurity: the Joinville DHA Clinical Trial

Published online by Cambridge University Press:  03 June 2024

Patrícia Zanotelli Cagliari*
Affiliation:
Postgraduate Program in Health and Environment, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil Darcy Vargas Maternity Hospital, Joinville, SC 89.202-190, Brazil Medicine Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil
Vinícius Ricardo Franzoi Hoeller
Affiliation:
Medicine Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil
Émelli Louise Rüncus Kanzler
Affiliation:
Medicine Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil
Melody Cristina Mansani Carraro
Affiliation:
Darcy Vargas Maternity Hospital, Joinville, SC 89.202-190, Brazil
Zaine Glaci Duarte Corrêa
Affiliation:
Nursing Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil
Gleici Blazius
Affiliation:
Nursing Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil
Pietra Giovanna Marghetti
Affiliation:
Nursing Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil
Gabriela Bruns Lenz
Affiliation:
Medicine Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil
Silmara Salete de Barros Silva Mastroeni
Affiliation:
Postgraduate Program in Health and Environment, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil Medicine Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil Nursing Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil Nutrition Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil
Marco Fabio Mastroeni
Affiliation:
Postgraduate Program in Health and Environment, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil Medicine Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil Nursing Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil Nutrition Department, University of Joinville Region – UNIVILLE, Joinville, SC 89.219-710, Brazil
*
*Corresponding author: Patricia Zanotelli Cagliari, email patriciacagliari@univille.br

Abstract

Retinopathy of prematurity (ROP) is a leading cause of blindness in premature infants. The condition is associated with DHA deficiency. This study aimed to investigate the effect of DHA supplementation on the occurrence of ROP in infants receiving oral oil drops. It is part of the Joinville DHA study, a non-parallel-group cohort study conducted from March 2020 to January 2023 at a public maternity hospital in Brazil. Infants born before 33 weeks of gestational age or with a birth weight ≤ 1500 g were recruited. Among 155 infants, 81 did not receive and 74 received DHA supplementation until complete vascularisation of the peripheral retina. There was a higher incidence of infants with ROP in the unsupplemented group (58·6 %) compared with the DHA group (41·4 %), but this difference was NS (P = 0·22). Unadjusted logistic regression analysis showed that patent ductus arteriosus and neonatal corticosteroids were significantly (P < 0·05) associated with ROP in both groups. In the DHA group, surfactant use was also associated with ROP (P = 0·003). After adjusting for important covariates, patent ductus arteriosus and neonatal corticosteroids continued to be significant for infants in the unsupplemented group (OR = 3·99; P = 0·022 and OR = 5·64; P = 0·019, respectively). In the DHA group, only surfactant use continued to be associated with ROP (OR = 4·84; P = 0·015). In summary, DHA supplementation was not associated with ROP. Further studies are necessary to better understand the relationship between DHA supplementation, ROP and associated comorbidities.

Type
Research Article
Copyright
© The Author(s), 2024. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Cagliari, PZ, Lucas, VC, Borba, IC, et al. (2019) Validation of ROPScore to predict retinopathy of prematurity among very low birth weight preterm infants in a southern Brazilian population. Arquivos Brasileiros Oftalmologia 82, 476480.CrossRefGoogle Scholar
Freitas, AM, Mörschbächer, R, Thorell, MR, et al. (2018) Incidence and risk factors for retinopathy of prematurity: a retrospective cohort study. Int J Retina Vitreous 4, 20.CrossRefGoogle ScholarPubMed
Gonçalves, E, Násser, LS, Martelli, DR, et al. (2014) Incidence and risk factors for retinopathy of prematurity in a Brazilian reference service. Sao Paulo Med J 132, 8591.CrossRefGoogle Scholar
Lermann, VL, Fortes Filho, JB & Procianoy, RS (2006) The prevalence of retinopathy of prematurity in very low birth weight newborn infants. J Pediatr (Rio J) 82, 2732.CrossRefGoogle ScholarPubMed
Goldstein, GP, Leonard, SA, Kan, P, et al. (2019) Prenatal and postnatal inflammation-related risk factors for retinopathy of prematurity. J Perinatol 39, 964973.CrossRefGoogle ScholarPubMed
Gonski, S, Hupp, SR, Cotten, CM, et al. (2019) Risk of development of treated retinopathy of prematurity in very low birth weight infants. J Perinatol 39, 15621568.CrossRefGoogle ScholarPubMed
Cestari, YLF, Lima, MAC, Rezende, ML, et al. (2021) Risk factors for retinopathy of prematurity: a systematic review. Revista Brasileira de Oftalmologia 80, 18.CrossRefGoogle Scholar
Filho, JBF, Bonomo, PP, Maia, M, et al. (2009) Weight gain measured at 6 weeks after birth as a predictor for severe retinopathy of prematurity: study with 317 very low birth weight preterm babies. Graefe’s Archive for Clin Exp Ophthalmol 247, 831836.CrossRefGoogle Scholar
Kim, SJ, Port, AD, Swan, R, et al. (2018) Retinopathy of prematurity: a review of risk factors and their clinical significance. Surv Ophthalmol 63, 618637.CrossRefGoogle ScholarPubMed
Kosmeri, C, Giapros, V, Gounaris, A, et al. (2023) Are the current feeding volumes adequate for the growth of very preterm neonates? Br J Nutr 130, 13381342.CrossRefGoogle ScholarPubMed
Stoltz Sjöström, E, Lundgren, P, Öhlund, I, et al. (2016) Low energy intake during the first 4 weeks of life increases the risk for severe retinopathy of prematurity in extremely preterm infants. Arch Dis Child Fetal Neonatal Ed 101, F108113.CrossRefGoogle ScholarPubMed
Bernabe-García, M, Villegas-Silva, R, Villavicencio-Torres, A, et al. (2019) Enteral docosahexaenoic acid and retinopathy of prematurity: a randomized clinical trial. JPEN J Parenter Enteral Nutr 43, 874882.CrossRefGoogle ScholarPubMed
Malamas, A, Chranioti, A, Tsakalidis, C, et al. (2017) The n-3 and retinopathy of prematurity relationship. Int J Ophthalmol 10, 300305.Google Scholar
Connor, WE, Neuringer, M & Reisbick, S (1992) Essential fatty acids: the importance of n-3 fatty acids in the retina and brain. Nutr Rev 50, 2129.CrossRefGoogle ScholarPubMed
Lapillonne, A & Moltu, SJ (2016) Long-chain polyunsaturated fatty acids and clinical outcomes of preterm infants. Ann Nutr Metab 69, 3544.CrossRefGoogle ScholarPubMed
Yang, R, Ding, H, Shan, J, et al. (2022) Association of fish oil containing lipid emulsions with retinopathy of prematurity: a retrospective observational study. BMC Pediatr 22, 113120.CrossRefGoogle ScholarPubMed
Najm, S, Löfqvist, C, Hellgren, G, et al. (2017) Effects of a lipid emulsion containing fish oil on polyunsaturated fatty acid profiles, growth and morbidities in extremely premature infants: a randomized controlled trial. Clin Nutr ESPEN 20, 1723.CrossRefGoogle ScholarPubMed
Pawlik, D, Lauterbach, R, Walczak, M, et al. (2014) 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 38, 711716.CrossRefGoogle ScholarPubMed
Hellström, A, Pivodic, A, Gränse, L, et al. (2021) Association of docosahexaenoic acid and arachidonic acid serum levels with retinopathy of prematurity in preterm infants. JAMA Netw Open 4, e2128771.CrossRefGoogle ScholarPubMed
Moltu, SJ, Bronsky, J, Embleton, N, et al. (2021) Nutritional management of the critically ill neonate: a position paper of the ESPGHAN committee on nutrition. J Pediatr Gastroenterol Nutr 73, 274289.CrossRefGoogle ScholarPubMed
Zin, A, Florêncio, T, Fortes Filho, JB, et al. (2007) Proposta de diretrizes brasileiras do exame e tratamento de retinopatia da prematuridade (ROP) (Brazilian guidelines proposal for screening and treatment of retinopathy of prematurity (ROP)). Arquivos Brasileiros Oftalmologia 70, 875883.CrossRefGoogle ScholarPubMed
Costa, MC, Eckert, GU, Fortes, BG, et al. (2013) Oral glucose for pain relief during examination for retinopathy of prematurity: a masked randomized clinical trial. Clinics (Sao Paulo) 68, 199204.CrossRefGoogle ScholarPubMed
Villar, J, Cheikh Ismail, L, Victora, CG, et al. (2014) International standards for newborn weight, length, and head circumference by gestational age and sex: the Newborn Cross-Sectional Study of the INTERGROWTH-21st Project. Lancet 384, 857868.CrossRefGoogle ScholarPubMed
Tomé, VAV, Vieira, JF, Oliveira, LB, et al. (2011) Estudo da retinopatia da prematuridade em um hospital universitário (Study of retinopathy of prematurity in a university hospital). Arquivos Brasileiros Oftalmologia 74, 279282.CrossRefGoogle ScholarPubMed
Collins, CT, Makrides, M, McPhee, AJ, et al. (2017) Docosahexaenoic acid and bronchopulmonary dysplasia in preterm infants. N Engl J Med 376, 12451255.CrossRefGoogle ScholarPubMed
Hellström, A, Nilsson, AK, Wackernagel, D, et al. (2021) Effect of enteral lipid supplement on severe retinopathy of prematurity: a randomized clinical trial. JAMA Pediatr 175, 359367.CrossRefGoogle ScholarPubMed
Wendel, K, Aas, MF, Gunnarsdottir, G, et al. (2023) Effect of arachidonic and docosahexaenoic acid supplementation on respiratory outcomes and neonatal morbidities in preterm infants. Clin Nutr 42, 2228.CrossRefGoogle ScholarPubMed
Beken, S, Dilli, D, Fettah, ND, et al. (2014) The influence of fish-oil lipid emulsions on retinopathy of prematurity in very low birth weight infants: a randomized controlled trial. Early Hum Dev 90, 2731.CrossRefGoogle ScholarPubMed
Frost, BL, Patel, AL, Robinson, DT, et al. (2021) Randomized controlled trial of early docosahexaenoic acid and arachidonic acid enteral supplementation in very low birth weight infants. J Pediatr 232, 2330.e21.CrossRefGoogle Scholar
Gillespie, TC, Kim, ES, Grogan, T, et al. (2022) Decreased levels of erythrocyte membrane arachidonic and docosahexaenoic acids are associated with retinopathy of prematurity. Invest Ophthalmol Vis Sci 63, 23.CrossRefGoogle ScholarPubMed
Pivodic, A, Johansson, H, Smith, LE, et al. (2022) Evaluation of the Retinopathy of Prematurity Activity Scale (ROP-ActS) in a randomised controlled trial aiming for prevention of severe ROP: a substudy of the Mega Donna Mega trial. BMJ Open Ophthalmol 7, e000923.CrossRefGoogle Scholar
Martin, CR (2014) Fatty acid requirements in preterm infants and their role in health and disease. Clin Perinatol 41, 363382.CrossRefGoogle ScholarPubMed
Löfqvist, CA, Najm, S, Hellgren, G, et al. (2018) Association of retinopathy of prematurity with low levels of arachidonic acid: a secondary analysis of a randomized clinical trial. JAMA Ophthalmol 136, 271277.CrossRefGoogle ScholarPubMed
Harris, WS & Baack, ML (2015) Beyond building better brains: bridging the docosahexaenoic acid (DHA) gap of prematurity. J Perinatol 35, 17.CrossRefGoogle ScholarPubMed
Ali, AA, Gomaa, NAS, Awadein, AR, et al. (2017) Retrospective cohort study shows that the risks for retinopathy of prematurity included birth age and weight, medical conditions and treatment. Acta Paediatr 106, 19191927.CrossRefGoogle ScholarPubMed
Porcelli, PJ & Weaver, RG Jr (2010) The influence of early postnatal nutrition on retinopathy of prematurity in extremely low birth weight infants. Early Hum Dev 86, 391396.CrossRefGoogle ScholarPubMed
Ho, MY & Yen, YH (2016) Trend of nutritional support in preterm infants. Pediatr Neonatol 57, 365370.CrossRefGoogle ScholarPubMed
Brown, JVE, Walsh, V & McGuire, W (2019) Formula v. maternal breast milk for feeding preterm or low birth weight infants. Cochrane Database Syst Rev 2019, issue 8, CD002972.Google ScholarPubMed
Connor, KM, SanGiovanni, JP, Lofqvist, C, et al. (2007) Increased dietary intake of n-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat Med 13, 868873.CrossRefGoogle Scholar
Barfield, WD (2018) Public health implications of very preterm birth. Clin Perinatol 45, 565577.CrossRefGoogle ScholarPubMed
Hamrick, SEG, Sallmon, H, Rose, AT, et al. (2020) Patent ductus arteriosus of the preterm infant. Pediatr 146, e20201209.CrossRefGoogle ScholarPubMed
Cavallaro, G, Filippi, L, Bagnoli, P, et al. (2014) The pathophysiology of retinopathy of prematurity: an update of previous and recent knowledge. Acta Ophthalmol 92, 220.CrossRefGoogle ScholarPubMed
Terrin, G, Di Chiara, M, Boscarino, G, et al. (2021) Morbidity associated with patent ductus arteriosus in preterm newborns: a retrospective case-control study. Ital J Pediatr 47, 9.CrossRefGoogle ScholarPubMed
Aydemir, O, Sarikabadayi, YU, Aydemir, C, et al. (2011) Adjusted poor weight gain for birth weight and gestational age as a predictor of severe ROP in VLBW infants. Eye (Lond) 25, 725729.CrossRefGoogle ScholarPubMed
Htun, ZT, Schulz, EV, Desai, RK, et al. (2021) Postnatal steroid management in preterm infants with evolving bronchopulmonary dysplasia. J Perinatol 41, 17831796.CrossRefGoogle ScholarPubMed
Buczynski, BW, Maduekwe, ET & O’Reilly, MA (2013) The role of hyperoxia in the pathogenesis of experimental BPD. Semin Perinatol 37, 6978.CrossRefGoogle ScholarPubMed
Mach, WJ, Thimmesch, AR, Pierce, JT, et al. (2011) Consequences of hyperoxia and the toxicity of oxygen in the lung. Nurs Res Pract 2011, 260482.Google ScholarPubMed
de Las Rivas Ramírez, N, Luque Aranda, G, Rius Díaz, F, et al. (2022) Risk factors associated with Retinopathy of Prematurity development and progression. Sci Rep 12, 21977.CrossRefGoogle ScholarPubMed
Trzcionkowska, K, Groenendaal, F, Andriessen, P, et al. (2021) Risk factors for retinopathy of prematurity in the Netherlands: a comparison of two cohorts. Neonatology 118, 462469.CrossRefGoogle ScholarPubMed
Doyle, LW (2021) Postnatal corticosteroids to prevent or treat bronchopulmonary dysplasia. Neonatology 118, 244251.CrossRefGoogle ScholarPubMed
Movsas, TZ, Spitzer, AR & Gewolb, IH (2016) Postnatal corticosteroids and risk of retinopathy of prematurity. J AAPOS 20, 348352.CrossRefGoogle ScholarPubMed
Kothadia, JM, O’Shea, TM, Roberts, D, et al. (1999) 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. Pediatr 104, 2227.CrossRefGoogle ScholarPubMed
Harmon, HM, Jensen, EA, Tan, S, et al. (2020) Timing of postnatal steroids for bronchopulmonary dysplasia: association with pulmonary and neurodevelopmental outcomes. J Perinatol 40, 616627.CrossRefGoogle ScholarPubMed
Marc, I, Piedboeuf, B, Lacaze-Masmonteil, T, et al. (2020) Effect of maternal docosahexaenoic acid supplementation on bronchopulmonary dysplasia-free survival in breastfed preterm infants: a randomized clinical trial. JAMA 324, 157167.CrossRefGoogle ScholarPubMed
Tanaka, K, Tanaka, S, Shah, N, et al. (2022) Docosahexaenoic acid and bronchopulmonary dysplasia in preterm infants: a systematic review and meta-analysis. J Matern Fetal Neonatal Med 35, 17301738.CrossRefGoogle ScholarPubMed
Freddi, NA, Filho, JO & Fiori, HH (2003) Exogenous surfactant therapy in pediatrics. J Pediatr (Rio J) 79, S205S212.CrossRefGoogle ScholarPubMed
Hentschel, R, Bohlin, K, van Kaam, A, et al. (2020) Surfactant replacement therapy: from biological basis to current clinical practice. Pediatr Res 88, 176183.CrossRefGoogle ScholarPubMed
Rebello, CM, Proença, RSM, Troster, EJ, et al. (2002) Terapia com surfactante pulmonar exógeno: o que é estabelecido e o que necessitamos determinar (Exogenous surfactant therapy – what is established and what still needs to be determined). Jornal de Pediatria 78, S215S226.Google Scholar
Janssen, CI & Kiliaan, AJ (2014) Long-chain polyunsaturated fatty acids (LCPUFA) from genesis to senescence: the influence of LCPUFA on neural development, aging, and neurodegeneration. Prog Lipid Res 53, 117.CrossRefGoogle ScholarPubMed
Saugstad, OD & Oei, JL, Lakshminrusimha, S, et al. (2019) Oxygen therapy of the newborn from molecular understanding to clinical practice. Pediatr Res 85, 2029.CrossRefGoogle ScholarPubMed
Soll, R & Ozek, E (2010) Prophylactic protein free synthetic surfactant for preventing morbidity and mortality in preterm infants. Cochrane Database Syst Rev 2010, issue 2010, CD001079.CrossRefGoogle ScholarPubMed
Supplementary material: File

Cagliari et al. supplementary material 1

Cagliari et al. supplementary material
Download Cagliari et al. supplementary material 1(File)
File 268.9 KB
Supplementary material: File

Cagliari et al. supplementary material 2

Cagliari et al. supplementary material
Download Cagliari et al. supplementary material 2(File)
File 25.2 KB