brand logo

EMMA J. PAR, MD, CHASE A. HUGHES, MD, AND PIERCE DERICO, MD

Am Fam Physician. 2023;107(5):525-534

Author disclosure: No relevant financial relationships.

Neonatal jaundice due to hyperbilirubinemia is common, and most cases are benign. The irreversible outcome of brain damage from kernicterus is rare (1 out of 100,000 infants) in high-income countries such as the United States, and there is increasing evidence that kernicterus occurs at much higher bilirubin levels than previously thought. However, newborns who are premature or have hemolytic diseases are at higher risk of kernicterus. It is important to evaluate all newborns for risk factors for bilirubin-related neurotoxicity, and it is reasonable to obtain screening bilirubin levels in newborns with risk factors. All newborns should be examined regularly, and bilirubin levels should be measured in those who appear jaundiced. The American Academy of Pediatrics (AAP) revised its clinical practice guideline in 2022 and reconfirmed its recommendation for universal neonatal hyperbilirubinemia screening in newborns 35 weeks' gestational age or greater. Although universal screening is commonly performed, it increases unnecessary phototherapy use without sufficient evidence that it decreases the incidence of kernicterus. The AAP also released new nomograms for initiating phototherapy based on gestational age at birth and the presence of neurotoxicity risk factors, with higher thresholds than in previous guidelines. Phototherapy decreases the need for an exchange transfusion but has the potential for short- and long-term adverse effects, including diarrhea and increased risk of seizures. Mothers of infants who develop jaundice are also more likely to stop breastfeeding, even though discontinuation is not necessary. Phototherapy should be used only for newborns who exceed thresholds recommended by the current AAP hour-specific phototherapy nomograms.

  • Immediate, unlimited access to all AFP content
  • More than 130 CME credits/year
  • AAFP app access
  • Print delivery available

Issue Access

  • Immediate, unlimited access to this issue's content
  • CME credits

Article Only

  • Immediate, unlimited access to just this article
  • American Academy of Pediatrics Subcommittee on Hyperbilirubinemia. Management of hyperbilirubinemia in the newborn infant 35 or more weeks of gestation [published correction appears in Pediatrics . 2004;114(4):1138]. Pediatrics. 2004;114(1):297-316.

Maisels MJ. Neonatal jaundice. Pediatr Rev. 2006;27(12):443-454.

Holland L, Blick K. Implementing and validating transcutaneous bilirubinometry for neonates. Am J Clin Pathol. 2009;132(4):555-561.

Kemper AR, Newman TB, Slaughter JL, et al. Clinical practice guideline revision: management of hyperbilirubinemia in the newborn infant 35 or more weeks of gestation. Pediatrics. 2022;150(3):e2022058859.

Gottesman LE, Del Vecchio MT, Aronoff SC. Etiologies of conjugated hyperbilirubinemia in infancy: a systematic review of 1692 subjects. BMC Pediatr. 2015;15:192.

Harpavat S, Garcia-Prats JA, Anaya C, et al. Diagnostic yield of newborn screening for biliary atresia using direct or conjugated bilirubin measurements. JAMA. 2020;323(12):1141-1150.

Davis AR, Rosenthal P, Escobar GJ, et al. Interpreting conjugated bilirubin levels in newborns. J Pediatr. 2011;158(4):562-565.e1.

Sgro M, Campbell D, Shah V. Incidence and causes of severe neonatal hyperbilirubinemia in Canada. CMAJ. 2006;175(6):587-590.

Usman F, Diala UM, Shapiro SM, et al. Acute bilirubin encephalopathy and its progression to kernicterus: current perspectives. Res Rep Neonatol. 2018;8:33-44.

Gamaleldin R, Iskander I, Seoud I, et al. Risk factors for neurotoxicity in newborns with severe neonatal hyperbilirubinemia. Pediatrics. 2011;128(4):e925-e931.

Moerschel SK, Cianciaruso LB, Tracy LR. A practical approach to neonatal jaundice. Am Fam Physician. 2008;77(9):1255-1262.

  • Sgro M, Campbell DM, Kandasamy S, et al. Incidence of chronic bilirubin encephalopathy in Canada, 2007–2008. Pediatrics. 2012;130(4):e886-e890.

Alkén J, Håkansson S, Ekéus C, et al. Rates of extreme neonatal hyper-bilirubinemia and kernicterus in children and adherence to national guidelines for screening, diagnosis, and treatment in Sweden. JAMA Netw Open. 2019;2(3):e190858.

Donneborg ML, Hansen BM, Vandborg PK, et al. Extreme neonatal hyperbilirubinemia and kernicterus spectrum disorder in Denmark during the years 2000–2015. J Perinatol. 2020;40(2):194-202.

Wu YW, Kuzniewicz MW, Wickremasinghe AC, et al. Risk for cerebral palsy in infants with total serum bilirubin levels at or above the exchange transfusion threshold: a population-based study. JAMA Pediatr. 2015;169(3):239-246.

US Preventive Services Task Force. Screening of infants for hyperbilirubinemia to prevent chronic bilirubin encephalopathy. Pediatrics. 2009;124(4):1172-1177.

Al-Lawama M, Badran E, Elrimawi A, et al. Intravenous immunoglobulins as adjunct treatment to phototherapy in isoimmune hemolytic disease of the newborn: a retrospective case-control study. J Clin Med Res. 2019;11(11):760-763.

Vidavalur R, Bhutani VK. Economic evaluation of point of care universal newborn screening for glucose-6-phosphate dehydrogenase deficiency in United States. J Matern Fetal Neonatal Med. 2022;35(25):5745-5753.

American Academy of Family Physicians. Infant jaundice. Updated October 5, 2020. Accessed March 8, 2023. https://familydoctor.org/condition/infant-jaundice

National Institute for Health and Care Excellence. Jaundice in newborn babies under 28 days. Clinical guideline [CG98]. Updated October 2016. Accessed March 3, 2023. https://www.nice.org.uk/guidance/cg98

Riskin A, Tamir A, Kugelman A, et al. Is visual assessment of jaundice reliable as a screening tool to detect significant neonatal hyperbilirubinemia?. J Pediatr. 2008;152(6):782-787.

Ostfeld-Johns S, Aragona E, Hart L. Removing race from hyperbilirubinemia guidelines is not enough. JAMA Pediatr. 2022;176(12):1163-1164.

Bhutani VK, Gourley GR, Adler S, et al. Noninvasive measurement of total serum bilirubin in a multiracial predischarge newborn population to assess the risk of severe hyperbilirubinemia. Pediatrics. 2000;106(2):E17.

Campbell DM, Danayan KC, McGovern V, et al. Transcutaneous bilirubin measurement at the time of hospital discharge in a multiethnic newborn population. Paediatr Child Health. 2011;16(3):141-145.

van den Esker-Jonker B, den Boer L, Pepping RMC, et al. Transcutaneous bilirubinometry in jaundiced neonates: a randomized controlled trial. Pediatrics. 2016;138(6):e20162414.

Mahram M, Oveisi S, Jaberi N. Transcutaneous bilirubinometery versus serum bilirubin in neonatal jaundice. Acta Med Iran. 2015;53(12):764-769.

Darling EK, Ramsay T, Sprague AE, et al. Universal bilirubin screening and health care utilization. Pediatrics. 2014;134(4):e1017-e1024.

Guidelines for detection, management and prevention of hyperbilirubinemia in term and late preterm newborn infants (35 or more weeks' gestation) - summary. Paediatr Child Health. 2007;12(5):401-418.

Kuzniewicz MW, Escobar GJ, Newman TB. Impact of universal bilirubin screening on severe hyperbilirubinemia and phototherapy use. Pediatrics. 2009;124(4):1031-1039.

Mah MP, Clark SL, Akhigbe E, et al. Reduction of severe hyperbilirubinemia after institution of predischarge bilirubin screening. Pediatrics. 2010;125(5):e1143-e1148.

Suresh GK, Clark RE. Cost-effectiveness of strategies that are intended to prevent kernicterus in newborn infants. Pediatrics. 2004;114(4):917-924.

Papacostas MF, Robertson DM, McLean MD, et al. Sixth-hour transcutaneous bilirubin and need for phototherapy in DAT positive infants. Pediatrics. 2022;149(3):e2021054071.

Kuzniewicz MW, Park J, Niki H, et al. Predicting the need for phototherapy after discharge. Pediatrics. 2021;147(5):e2020019778.

Lauer BJ, Spector ND. Hyperbilirubinemia in the newborn. Pediatr Rev. 2011;32(8):341-349.

Chang PW, Waite WM. Evaluation of home phototherapy for neonatal hyperbilirubinemia. J Pediatr. 2020;220:80-85.

Chu L, Qiao J, Xu C. Home-based phototherapy versus hospital-based phototherapy for treatment of neonatal hyperbilirubinemia: a systematic review and meta-analysis. Clin Pediatr (Phila). 2020;59(6):588-595.

Kumar P, Chawla D, Deorari A. Light-emitting diode phototherapy for unconjugated hyperbilirubinaemia in neonates. Cochrane Database Syst Rev. 2011(12):CD007969.

Muchowski KE. Evaluation and treatment of neonatal hyperbilirubinemia. Am Fam Physician. 2014;89(11):873-878.

Newman TB, Wu YW, Kuzniewicz MW, et al. Childhood seizures after phototherapy. Pediatrics. 2018;142(4):e20180648.

Maimburg RD, Olsen J, Sun Y. Neonatal hyperbilirubinemia and the risk of febrile seizures and childhood epilepsy. Epilepsy Res. 2016;124:67-72.

Wickremasinghe AC, Kuzniewicz MW, Grimes BA, et al. Neonatal phototherapy and infantile cancer. Pediatrics. 2016;137(6):e20151353.

Newman TB, Wickremasinghe AC, Walsh EM, et al. Retrospective cohort study of phototherapy and childhood cancer in Northern California. Pediatrics. 2016;137(6):e20151354.

Auger N, Laverdière C, Ayoub A, et al. Neonatal phototherapy and future risk of childhood cancer. Int J Cancer. 2019;145(8):2061-2069.

Digitale JC, Kim MO, Kuzniewicz MW, et al. Update on phototherapy and childhood cancer in a Northern California cohort. Pediatrics. 2021;148(5):e2021051033.

Wickremasinghe AC, Kuzniewicz MW, McCulloch CE, et al. Efficacy of subthreshold newborn phototherapy during the birth hospitalization in preventing readmission for phototherapy. JAMA Pediatr. 2018;172(4):378-385.

Besser I, Perry ZH, Mesner O, et al. Yield of recommended blood tests for neonates requiring phototherapy for hyperbilirubinemia. Isr Med Assoc J. 2010;12(4):220-224.

Willis SK, Hannon PR, Scrimshaw SC. The impact of the maternal experience with a jaundiced newborn on the breastfeeding relationship. J Fam Pract. 2002;51(5):465.

Kemper K, Forsyth B, McCarthy P. Jaundice, terminating breast-feeding, and the vulnerable child. Pediatrics. 1989;84(5):773-778.

Pace EJ, Brown CM, DeGeorge KC. Neonatal hyperbilirubinemia: an evidence-based approach. J Fam Pract. 2019;68(1):E4-E11.

Ip S, Chung M, Kulig J, et al.; American Academy of Pediatrics Subcommittee on Hyperbilirubinemia. An evidence-based review of important issues concerning neonatal hyperbilirubinemia. Pediatrics. 2004;114(1):e130-e153.

Jackson JC. Adverse events associated with exchange transfusion in healthy and ill newborns. Pediatrics. 1997;99(5):E7.

Grabenhenrich J, Grabenhenrich L, Büher C, et al. Transcutaneous bilirubin after phototherapy in term and preterm infants. Pediatrics. 2014;134(5):e1324-e1329.

  • Berkwitt A, Osborn R, Grossman M. The utility of inpatient rebound bilirubin levels in infants readmitted after birth hospitalization for hyperbilirubinemia. Hosp Pediatr. 2015;5(2):74-78.

Flaherman VJ, Maisels MJ; Academy of Breastfeeding Medicine. ABM clinical protocol #22: guidelines for the management of jaundice in the breastfeeding infant 35 weeks or more gestation – revised 2017. Breastfeed Med. 2017;12(5):250-257.

Porter ML, Dennis BL. Hyperbilirubinemia in the term newborn. Am Fam Physician. 2002;65(4):599-606.

Continue Reading

case study neonatal hyperbilirubinemia

More in AFP

More in pubmed.

Copyright © 2023 by the American Academy of Family Physicians.

This content is owned by the AAFP. A person viewing it online may make one printout of the material and may use that printout only for his or her personal, non-commercial reference. This material may not otherwise be downloaded, copied, printed, stored, transmitted or reproduced in any medium, whether now known or later invented, except as authorized in writing by the AAFP.  See permissions  for copyright questions and/or permission requests.

Copyright © 2024 American Academy of Family Physicians. All Rights Reserved.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

Risk factors for severe hyperbilirubinemia in neonates

Affiliation.

  • 1 Department of Medical Technology, Fooyin University, 151 Chin-Hsueh Rd., Ta-Liao Hsiang, Kaohsiung Hsien 831, Taiwan. [email protected]
  • PMID: 15319464
  • DOI: 10.1203/01.PDR.0000141846.37253.AF

The incidence of severe neonatal hyperbilirubinemia is higher in Asians than in whites. A case-control study was designed to investigate the effects of eight known risk factors [breast feeding, ABO incompatibility, premature birth, infection, cephalohematoma, asphyxia, glucose-6-phosphate dehydrogenase (G6PD) deficiency, and variant UDP-glucuronosyltransferase 1A1 (UGT1A1) gene] and a suspicious analog [organic anion transporter 2 (OATP 2) gene] on severe hyperbilirubinemia in Taiwanese neonates. The 72 study subjects and 100 hospital control subjects consisted of neonates with peak serum bilirubin levels > or =342 microM and <256.5 microM, respectively. The PCR-restriction fragment length polymorphism method was applied to detect the UGT1A1, OATP 2, and G6PD genes. The results of multivariate logistic regressions, adjusted for covariates, revealed odds ratios (ORs) of 4.64 [95% confidence interval (CI): 2.25-9.57; p < 0.001], 3.36 (95% CI: 1.54-7.35; p=0.002), and 3.02 (95% CI: 1.30-6.99; p=0.010) for neonates who were fed with breast milk, and carry the variant UGT1A1 gene at nucleotide 211 and the variant OATP 2 gene at nucleotide 388, respectively. The ORs, adjusted for covariates, for the other six risk factors were not statistically significant. The ORs in neonates who had one, two, and three significant risk factors were 8.46 (95% CI: 2.75-34.48; p < 0.001), 22.0 (95% CI: 5.50-88.0; p < 0.001), and 88.0 (95% CI: 12.50-642.50; p < 0.001), respectively. In conclusion, neonates who carry the 211 and 388 variants in the UGT1A1 and OATP 2 genes, respectively, as well as feed with breast milk are at high risk to develop severe hyperbilirubinemia.

PubMed Disclaimer

  • Genetics and the risk of neonatal hyperbilirubinemia: commentary on the article by Huang et al. on page 682. Watchko JF. Watchko JF. Pediatr Res. 2004 Nov;56(5):677-8. doi: 10.1203/01.PDR.0000142588.65045.25. Epub 2004 Sep 15. Pediatr Res. 2004. PMID: 15371564 Review. No abstract available.

Similar articles

  • Identifying term breast-fed infants at risk of significant hyperbilirubinemia. Chang PF, Lin YC, Liu K, Yeh SJ, Ni YH. Chang PF, et al. Pediatr Res. 2013 Oct;74(4):408-12. doi: 10.1038/pr.2013.120. Epub 2013 Jul 15. Pediatr Res. 2013. PMID: 23857295
  • Risk of hyperbilirubinemia in breast-fed infants. Chang PF, Lin YC, Liu K, Yeh SJ, Ni YH. Chang PF, et al. J Pediatr. 2011 Oct;159(4):561-5. doi: 10.1016/j.jpeds.2011.03.042. Epub 2011 May 18. J Pediatr. 2011. PMID: 21592495
  • Exploring the genetic architecture of neonatal hyperbilirubinemia. Watchko JF, Lin Z. Watchko JF, et al. Semin Fetal Neonatal Med. 2010 Jun;15(3):169-75. doi: 10.1016/j.siny.2009.11.003. Epub 2009 Dec 21. Semin Fetal Neonatal Med. 2010. PMID: 20022574 Review.
  • Complex multifactorial nature of significant hyperbilirubinemia in neonates. Watchko JF, Lin Z, Clark RH, Kelleher AS, Walker MW, Spitzer AR; Pediatrix Hyperbilirubinemia Study Group. Watchko JF, et al. Pediatrics. 2009 Nov;124(5):e868-77. doi: 10.1542/peds.2009-0460. Epub 2009 Oct 26. Pediatrics. 2009. PMID: 19858149
  • Molecular genetics of unconjugated hyperbilirubinemia in Taiwanese. Huang CS. Huang CS. J Biomed Sci. 2005;12(3):445-50. doi: 10.1007/s11373-005-3863-5. J Biomed Sci. 2005. PMID: 15965581 Review.
  • Do Gene Polymorphisms Play a Role in Newborn Hyperbilirubinemia? Hakan N, Aydin M, Ceylaner S, Dilli D, Zenciroğlu A, Okumuş N. Hakan N, et al. Balkan J Med Genet. 2024 Mar 12;26(2):51-58. doi: 10.2478/bjmg-2023-0021. eCollection 2023 Dec. Balkan J Med Genet. 2024. PMID: 38482260 Free PMC article.
  • Factors associated with neonatal jaundice among neonates admitted at referral hospitals in northeast Ethiopia: a facility-based unmatched case-control study. Ayalew T, Molla A, Kefale B, Alene TD, Abebe GK, Ngusie HS, Zemariam AB. Ayalew T, et al. BMC Pregnancy Childbirth. 2024 Feb 21;24(1):150. doi: 10.1186/s12884-024-06352-y. BMC Pregnancy Childbirth. 2024. PMID: 38383399 Free PMC article. Review.
  • Health seeking behaviour and knowledge on neonatal danger signs among neonatal caregivers in Upper Denkyira East Municipality, Ghana. Gyaase P, Aduse-Poku E, Lanquaye MO, Acheampong EB, Sampson DB. Gyaase P, et al. BMC Pediatr. 2024 Jan 8;24(1):27. doi: 10.1186/s12887-023-04430-2. BMC Pediatr. 2024. PMID: 38191444 Free PMC article.
  • The Genetics of Glucose-6-Phosphate-Dehydrogenase (G6PD) and Uridine Diphosphate Glucuronosyl Transferase 1A1 (UGT1A1) Promoter Gene Polymorphism in Relation to Quantitative Biochemical G6PD Activity Measurement and Neonatal Hyperbilirubinemia. Riskin A, Bravdo Y, Habib C, Maor I, Mousa J, Shahbarat S, Shahak E, Shalata A. Riskin A, et al. Children (Basel). 2023 Jul 6;10(7):1172. doi: 10.3390/children10071172. Children (Basel). 2023. PMID: 37508669 Free PMC article.
  • Contribution of genetic factors to high rates of neonatal hyperbilirubinaemia on the Thailand-Myanmar border. Bancone G, Gornsawun G, Peerawaranun P, Penpitchaporn P, Paw MK, Poe DD, Win D, Cicelia N, Mukaka M, Archasuksan L, Thielemans L, Nosten F, White NJ, McGready R, Carrara VI. Bancone G, et al. PLOS Glob Public Health. 2022 Jun 17;2(6):e0000475. doi: 10.1371/journal.pgph.0000475. eCollection 2022. PLOS Glob Public Health. 2022. PMID: 36962413 Free PMC article.

Publication types

  • Search in MeSH

Related information

  • Gene (GeneRIF)
  • Nucleotide (RefSeq)
  • Protein (RefSeq)
  • PubChem Compound
  • PubChem Substance

LinkOut - more resources

Full text sources.

  • Nature Publishing Group
  • Ovid Technologies, Inc.

Other Literature Sources

  • The Lens - Patent Citations

Miscellaneous

  • NCI CPTAC Assay Portal

full text provider logo

  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Review Article
  • Published: 04 May 2021

Diagnostic methods for neonatal hyperbilirubinemia: benefits, limitations, requirements, and novel developments

  • Christian V. Hulzebos 1 ,
  • Libor Vitek 2 ,
  • Carlos D. Coda Zabetta 3 ,
  • Aleš Dvořák 2 ,
  • Paul Schenk 4 ,
  • Eline A. E. van der Hagen 5 , 6 ,
  • Christa Cobbaert 4 &
  • Claudio Tiribelli 7  

Pediatric Research volume  90 ,  pages 277–283 ( 2021 ) Cite this article

3483 Accesses

21 Citations

1 Altmetric

Metrics details

Invasive bilirubin measurements remain the gold standard for the diagnosis and treatment of infants with severe neonatal hyperbilirubinemia. The present paper describes different methods currently available to assess hyperbilirubinemia in newborn infants. Novel point-of-care bilirubin measurement methods, such as the BiliSpec and the Bilistick, would benefit many newborn infants, especially in low-income and middle-income countries where the access to costly multi-analyzer in vitro diagnostic instruments is limited. Total serum bilirubin test results should be accurate within permissible limits of measurement uncertainty to be fit for clinical purposes. This implies correct implementation of internationally endorsed reference measurement systems as well as participation in external quality assessment programs. Novel analytic methods may, apart from bilirubin, include the determination of bilirubin photoisomers and bilirubin oxidation products in blood and even in other biological matrices.

Key message: Bilirubin measurements in blood remain the gold standard for diagnosis and treatment of severe neonatal hyperbilirubinemia (SNH). External quality assessment (EQA) plays an important role in revealing inaccuracies in diagnostic bilirubin measurements.

What does this article add to the existing literature? We provide analytic performance data on total serum bilirubin (TSB) as measured during recent EQA surveys. We review novel diagnostic point-of-care (POC) bilirubin measurement methods and analytic methods for determining bilirubin levels in biological matrices other than blood.

Impact: Manufacturers should make TSB test results traceable to the internationally endorsed total bilirubin reference measurement system and should ensure permissible limits of measurement uncertainty.

You have full access to this article via your institution.

Similar content being viewed by others

case study neonatal hyperbilirubinemia

Large scale validation of a new non-invasive and non-contact bilirubinometer in neonates with risk factors

case study neonatal hyperbilirubinemia

Screening methods for neonatal hyperbilirubinemia: benefits, limitations, requirements, and novel developments

case study neonatal hyperbilirubinemia

Natural history of early neonatal bilirubinemia: a global perspective

Introduction.

International guidelines for the management of neonates with unconjugated hyperbilirubinemia include treatment thresholds that are based on total serum bilirubin (TSB) concentrations. Bilirubin measurements are key to the management of neonatal jaundice. An essential prerequisite for bilirubin measurements is that they are accurate and precise to manage jaundiced newborn infants appropriately. Over the past decades, bilirubin measurement for severe neonatal hyperbilirubinemia (SNH) identification has constituted a major challenge. A vast number of methods to determine bilirubin in human serum have been developed since it was first reported in 1858 by Frerichs (Gmelin reaction). 1 , 2 Van den Bergh and Snapper described their important diazo reaction in 1913 3 and the colorimetric determination by Malloy and Evelyn was published in 1937. 4 This was followed by Jendrassik and Grof who refined and modified the diazo reactions in 1938. 5 For a thorough understanding in the subsequent paragraphs of the so-called science of measurement and of terms such as TSB accuracy, trueness, metrological traceability, and measurement uncertainty, we refer to the definitions and terminology provided by the International Vocabulary of Metrology. 6

Diagnostic methods—invasive total serum bilirubin measurements

Multiparameter instruments.

Routine laboratory bilirubin measurement is commonly performed with multichannel instruments that provide values for conjugated (direct), unconjugated (indirect), and TSB concentrations. They usually base the direct determination of bilirubin on diazo (Jendrassik Grof) and vanadate oxidase chemical reactions, or on variants. 7 On account of the high costs involved and the requirement of specialized personnel, these instruments are used mainly in laboratories of large hospitals. Even though the concentrations supplied by these instruments were considered as the clinical “reference” for TSB, inconsistencies amongst the different laboratory methods have been observed for decades. 8 , 9 In 2010, in the Netherlands, Cobbaert and colleagues analyzed the accuracy of TSB levels nationwide as measured by the most commonly used multiparameter instruments and in vitro diagnostic devices (IVDs). 10 In this study, a pooled human serum was supplemented with unconjugated bilirubin to obtain target values of 26.7 µmol/L (95% confidence interval (CI) range of 26.1–27.3 µmol/L), and 68.7 µmol/L (95% CI range of 67.2–70.2 µmol/L), as assigned with the Doumas reference measurement procedure (RMP) of the Joint Committee for Traceability in Laboratory Medicine (JCTLM) listed reference laboratory in Hannover, Germany. The Doumas RMP can be considered as the gold standard. The two value-assigned specimens were measured in 183 medical laboratories and in-house by IVD manufacturers using their respective routine methods. This procedure allows the accuracy of results produced by Dutch medical laboratories and IVD manufacturers to be assessed and to be compared to the target values assigned with an internationally recognized reference method. The interlaboratory variability and inaccuracy of TSB levels observed among manufacturers and individual laboratories were substantial. This indicates that the concept of metrological traceability, which leads to exchangeable TSB results, was not uniformly adopted in the commercially available IVDs. Also, in-house results produced by individual IVD manufacturers demonstrated significant differences. Similar discrepancies were observed by Greene and colleagues when comparing the performance of a Beckman AU680 instrument versus Ortho-Clinical-Diagnostics’ VITROS device. 11

It is common in neonatal intensive care units (NICUs) to find blood gas analyzer instruments like Radiometer ABL models or the RAPIDPoint models produced by Siemens HealthCare Diagnostics Inc. 12 Blood gas analyzer instruments estimate TSB indirectly by using whole blood co-oximetry, whereby whole blood is hemolyzed and serum equivalent bilirubin concentration calculated. Previously, the comparability of blood gas analysis-derived TSB levels and TSB levels measured on routine laboratory instruments was assessed. In 2018, Lano and colleagues reported a comparative analysis of neonatal TSB levels by whole blood co-oximetry (Radiometer® ABL90) against plasma bilirubin methods (Roche Diagnostics Cobas® C-601 and Ortho Clinical Diagnostics VITROS® 350). 13 Results showed good correlation in comparison to the Roche plasma diazo method, with a mean bias of −1.0 µmol/L across the bilirubin range examined and a 95% confidence interval range of −20.00 to 19.00 μmol/L. However, a statistically significant underestimation was found against the VITROS® 350 machine with a mean bias of –4.4 µmol/L over the bilirubin range examined and a 95% confidence interval of –29.90 to 21.10 μmol/L. Performance of the GEM Premier 4000® blood gas analyzer (Instrumentation Laboratory, Bedford, MA) was also assessed showing a wide range of differences compared to VITROS, with a negative bias at low concentrations of bilirubin and a positive bias at higher concentrations. Moreover, hemoglobin concentration and hemolysis affected the recovery of the GEM blood gas analyzer results. 14

Bench-top bilirubinometers

Other commonly used instruments for TSB measurement are bench-top bilirubinometers. Based on direct spectrophotometry, these are simple and rapid alternatives for assessing TSB that require a minimal sample for analysis. In practice, an undiluted serum is used to measure the bilirubin absorbance (at 454 nm) and hemoglobin (at 454 and 528 nm). Hence, subtracting the absorbance at 528 nm from that at 454 nm yields a value that can be attributed largely to bilirubin. The prevalence of the other forms of bilirubin and chromophores in older children and adults limits the use of this technique to neonates younger than 2–3 weeks of age. 15 Two types of direct spectrometry instruments are available commercially: those using sample cuvettes such as UNISTAT (Reichert Technologies, USA), and those using hematocrit capillary tubes like One Beam (Ginevri, Italy). Although there are many options available commercially of instruments based on direct spectrophotometry, validation studies of this method are limited. The advantage of bilirubinometers is the short turnaround times for results as shown in Table  1 . The requirement of sample processing, however, and the need for additional instrumentation such as centrifuge and trained laboratory personnel limits the use of this method for TSB determination substantially. 16 , 17

Hand-held point-of-care (POC) bilirubin instruments

Neonatal jaundice identification has always posed a challenge, mainly in LMICs. 18 , 19 Over the last years, with the advancement of technology, different solutions have emerged for SNH screening. In 2017, Keahey and colleagues reported validation data of a new screening device under development, the BiliSpec, in 94 blood samples of 67 newborn infants between the day of birth and 24 days. 20 This screening method is based on a battery-powered low-cost reader designed to quantify serum bilirubin levels from whole blood applied to a lateral flow card. 20 From a maintenance point of view, a drawback is that this device requires daily calibration for routine use. The study showed a high correlation ( r  = 0.97) of BiliSpec against a bench-top bilirubinometer (UNISTAT; Reichert Technologies) within a TSB range from 19 to 393 µmol/L with a mean (± SD)  value of 181 ± 68 µmol/L and differences up to 51 µmol/L (68% of total samples deviated ≤ 17 µmol/L). The mean bias between bench-top TSB and BiliSpec bilirubin readings was 5 µmol/L, with 95% limits of agreement of −29 to 38 µmol/L. In practice, lateral flow cards are designed to accept drops of whole blood obtained directly from a heel or finger prick. The separation from corpuscular components of the blood allows the flow of plasma into the nitrocellulose (NC) membrane by capillarity. Once the operator has visually interpreted that the NC membrane is saturated, the card is inserted into the reader for bilirubin measurement. The authors report that the design of the card allows controlling the volume of blood applied. The variability, however, in bilirubin test results could be the effect of an undersaturation or oversaturation of the NC membrane. In recent years, TSB measurement by another POC diagnostic method on capillary or venous blood samples became available with the Bilistick® System 1.0 (BM-BS 1.0 - Bilimetrix, Italy). 21 This direct method consists of a hand-held, rechargeable battery reflectance reader and test strips composed of a blood plasma separator coupled with an NC membrane, both encased in a plastic cassette. After loading the whole blood sample on the strip (35 µL - hematocrit up to 70%), it requires less than 100 s for serum separation and NC membrane saturation, depending on the hematocrit of the sample (identified automatically by the reader using light reflectance measurements to detect serum flow stabilization). The reader measures reflected light from the plasma-saturated NC membrane, using a light-emitting diode (LED) with an emission peak at 465 nm for quantifying bilirubin. A second LED of 570 nm detects whether hemoglobin contamination is present. The instrument is internally calibrated to optimize sensitivity and provides TSB measuring in a range of 17 to 684 µmol/L. The accuracy of the Bilistick® System 1.0 device was documented by comparing results with TSB measurements from routine laboratories. In 2018, Greco and colleagues reported the performance of the Bilistick® System 1.0 for identifying SNH in a multi-country approach in 1911 newborns. They showed that the TSB level measured by Bilistick® System 1.0 was not significantly different from laboratory TSB values in all four countries. 22 The Bilistick® System had a positive predictive value (PPV) of 92.5% and a negative predictive value (NPV) of 92.8%. When Greco and colleagues compared the Bilistick® System 1.0 with both transcutaneous bilirubinometry (JM-103) and laboratory TSB results (Synchron CX PRU 16360, Beckman-Coulter, USA), they found the Bilistick® System 1.0 to be a good alternative to transcutaneous bilirubin determination for early diagnosis and proper management of neonatal jaundice. 23 In 2018, Thielemans and colleagues reported a rather high failure rate for the Bilistick® System 1.0, especially in highly humid climatic conditions and at high hematocrit values. 24 In 2020, Kamineni and colleagues called to further improve the accuracy of the Bilistick. 25 Despite of these observations, the reliability and clinical use of the Bilistick® System 1.0 for measuring TSB was considered to be appropriate in other studies performed under similar weather conditions. 22 , 26 , 27 POC diagnosis of hyperbilirubinemia has also been claimed by Tabatabaee and colleagues who reported fast and reproducible TSB measurements in whole blood with a recently developed smartphone-based bilirubin assay kit using photoluminescent bacterial cellulose nanopaper. 28 One of the main advantages of the portable POC bilirubin instruments is the much shorter turnaround time, that is the interval between collecting the specimen and reporting the TSB result, compared to commonly used multi-analyzer instruments for TSB tests (Table  1 ). 27 Low-cost POC instruments appear to be an effective alternative for the measurement of TSB in newborns, particularly when conventional laboratory methods are not available or inaccessible.

As shown by comparative analysis, the unacceptable high variability in TSB measurement among methods continues to pose a real challenge to result harmonization of clinical routine methods. When Lo and colleagues evaluated the trueness of neonatal TSB using value-assigned, commutable specimens in four major instrument groups (Dimension, Olympus, Synchron, and VITROS), they found a systematic error in TSB measurement associated primarily with the failure of instrument manufacturers to produce reliable bilirubin calibrators. 17 High variabilities were also observed when comparing TSB levels in patient specimens on multiparameter analyzers, transcutaneous bilirubin, and direct spectrophotometry instruments. 29 Other potential sources of inaccuracy include sample integrity and sample handling. These were, however, typically identified as random errors. An in-depth analysis of results reported by the College of American Pathologists Neonatal Bilirubin PT Survey from 2011 to 2015, showed how changes in TSB test results—when trying to recalibrate instruments—can lead to completely opposite clinical interpretations. 30 Standardization of TSB measurement remains a formidable challenge for laboratory medicine. 31 , 32

The relevance of correctly implementing the internationally endorsed reference measurement systems

Inaccuracy and non-equivalence of TSB results among IVD manufacturers are well known. 17 , 30 But why are TSB tests not standardized adequately? It appears that there is insufficient awareness of the metrological traceability concept and its essential implementation through an unbroken chain of calibration hierarchies. Standardization is key to guaranteeing that TSB test results correspond properly to internationally agreed standards of a higher order (Fig.  1 ). 33 , 34 To achieve global standardization of measurement results in medical laboratories, the International Federation of Clinical Chemistry and Laboratory Medicine (IFCC) and the JCTLM promote the concept of metrological traceability of test results to internationally accepted standards. For proper commercial test calibration, medical test results must be anchored to higher-order reference materials and higher-order reference procedures. In the case of total bilirubin, tests were standardized with a Doumas reference method for bilirubin (using diazo-based spectrophotometry) and National Institute of Standards and Technology Standard Reference Material (NIST SRM) 916a reference material according to a strict calibration hierarchy. The former NIST SRM 916a reference material went missing years ago (it ran out of stock) and consisted of three isomers, two of which were not present in native human sera. It was decided therefore to use the specific molar extinction coefficient ( ε ) of 7649 m 2 /mol for bilirubin quantitation as it is superior to control the accuracy of a standard solution. Consequently, an updated and extended reference measurement system (RMS) was established and has been in place since 2018. To that end, Klauke and colleagues re-evaluated the Doumas candidate Reference Method and established a next-generation RMS without any need for a calibrator or SRM. 35 Currently, in this RMS total bilirubin is described as a so-called operative measurand, defined by a set of measurement parameters.

figure 1

This measurement system was adapted from ISO 17511:2020 and based on the re-evaluated reference measurement procedure from Klauke and colleagues. 35 Total serum bilirubin is described as an operative measurand, as defined by the reference measurement procedure. Through this reference measurement system, with its unbroken chain of calibrator materials and methods, bilirubin test results are anchored and made traceable to the International System of Units. Measurement uncertainty should remain within allowable limits to make the test fit for the purpose. SI International System of Units; ε the molar absorption coefficient of bilirubin (conventional quantity value of 7649 m 2 /mol); Mfr, IVD manufacturer who supplies CE-IVD kits for routine analyses to the European market.

The German Society for Clinical Chemistry and Laboratory Medicine runs the Reference Laboratories International Federation of Clinical Chemistry (RELA IFCC) external quality assessment scheme. It periodically checks how to reference laboratories perform using their RMP, i.e., the gold standard for total bilirubin ( http://www.dgkl-rfb.de:81 ).

Notwithstanding the existence of approved reference measurement systems (the former Doumas and AACC RMS (1985) versus the new RMS (2018)), 35 the levels of standardization and trueness of routine total bilirubin test results consistently differ amongst manufacturers—as demonstrated in external quality assessment (EQA) programs that use commutable EQA samples—and has often been questioned. Yet, TSB tests can only be declared fit for clinical purposes if results are accurate within permissible limits of measurement uncertainty. The latter is often translated into desirable analytic performance specifications derived from biological variation data. In the case of TSB, rational and desirable analytic performances in adults are a CV a (analytical variation) of <12.8%, an allowable bias of less than 10%, and a total allowable error of 31.1% ( https://biologicalvariation.eu/ ). 36 For neonatal bilirubin no biological variation data are available. Consequently, pediatricians can determine the analytic performance criteria needed to make the neonatal bilirubin test fit for purpose themselves.

Relevance of accuracy-based external quality assessment for trueness verification

External quality assessment (EQA) plays an essential role in helping to assure the quality of laboratory medicine on a daily basis. EQA schemes may reveal significant and systematic between-method variability for measurements of the same analyte in the same specimen. Detection of between-method variability through EQA is also a major driver for further improvement of test standardization.

Medical laboratories are obliged to perform an EQA and when used effectively it can provide many opportunities for improving test accuracy. In the Netherlands, an accuracy-based EQA was developed by the Dutch Foundation for Quality Assessment in Medical Laboratories (Stichting Kwaliteitsbewaking Medische Laboratoria, SKML) for general chemistry analytes. Thus far, however, these did not include TSB. To develop an accuracy-based EQA, native, commutable, value-assigned EQA samples are essential to give medical laboratories insight into trueness and imprecision of their bilirubin tests as well as other chemistry tests. In this way, inaccuracy, and absolute bias as a result of, for example, lot-to-lot variation or method changes, can be monitored in a longitudinal and sustainable way by every participating laboratory. To date, no proven commutable EQA samples have been developed for TSB. This has not been accomplished yet because the EQA samples used until now are spiked with either unconjugated and/or conjugated bilirubin. It was demonstrated that the TSB recoveries are affected by the spiking material, hampering trueness verification. Nevertheless, interlaboratory and intermethod variations are monitored bi-weekly for TSB in approximately 185 Dutch laboratories. Figure  2 demonstrates the interlaboratory and intermethod variations in Dutch medical laboratories using common reagents. In the recent EQA surveys, SKML 2019.4 and SKML 2020.1, overall interlaboratory CVs ranged from 3% to 6% for TSB in the concentration range of 13 to 110 µmol/L. The interlaboratory spreading has improved compared to the situation in 2009 (data not shown).

figure 2

Pooled human serum was supplemented with bilirubin. Serum samples were dispensed, frozen below −70 °C, and shipped on dry ice to the laboratories participating in the regular general clinical chemistry external quality assessment program of the Dutch Foundation for Quality Assessment in Medical Laboratories (SKML). In 2009 ( a ) and 2020 ( c ), samples were supplemented with unconjugated bilirubin alone (>98%, mixed isomers, Sigma-Aldrich). In 2019 ( b ), both unconjugated bilirubin and conjugated bilirubin were added. Each participating laboratory-measured total bilirubin. Results are plotted with the high-concentration sample on the x axis and the low-concentration sample on the y axis (17.1 µmol/L = 1 mg/dL). Colored squares represent mean bilirubin concentrations ± 1 standard deviation in both samples, as measured by the routine medical laboratories in the quality assessment program and using the methods by the respective manufacturers as indicated. The numbers inside the colored squares indicate the numbers of participants for each category. In panel a , stars represent bilirubin concentrations measured in-house by the corresponding manufacturers. The red dots are the targets set in the JCTLM-listed reference laboratory of Prof. Gerhard Schumann and Dr. Denis Grote-Koska in Hannover, Germany (Institut für Klinische Chemie - Zentrallabor, Medizinische Hochschule Hannover). Panel a was adapted from Cobbaert and colleagues. 10 Data for panels b and c were supplied by the Dutch Foundation for Quality Assessment in Medical Laboratories and used with their permission.

Other methods to study bilirubin metabolism—determination of bilirubin in tissues and cells

It is well known that the concentration of unconjugated free bilirubin correlates better with bilirubin neurotoxicity in comparison to total serum bilirubin concentration. Because bilirubin behaves like a real signaling molecule, 37 its blood concentrations can serve only as a rough surrogate marker of bilirubin metabolism within the cells. Although serum or plasma bilirubin concentrations are in dynamic equilibrium with other biological compartments, its tissue and cell concentrations differ substantially even within one organ as proved, for example, in the brain. 38 Knowledge of bilirubin kinetics and dynamics within individual human body compartments is essential to understand its role in the pathophysiology of various clinical conditions. Besides, bilirubin undergoes extensive metabolization, in particular, due to oxidation and photooxidation processes, forming tetra-, tri-, di- as well as monopyrrolic oxidation derivatives, which are likely to exert biological activities and may serve as important biomarkers of pathological conditions. Because cellular bilirubin concentrations are within the submicromolar range, the standard analytic, mostly diazo reaction-based methods, which are used in routine clinical chemistry, have insufficient sensitivity, and do not enable quantitation of bilirubin in the cells, tissues, and organs. This drawback of standard clinical chemistry methods is eliminated by using high-performance liquid chromatography (HPLC) techniques. These techniques enable accurate separation and quantitation of individual bilirubin fractions, such as delta bilirubin, unconjugated bilirubin, bilirubin monoglucuronosides, and diglucuronosides, and can be used under specific circumstances even in clinical settings. 39 Simultaneously, HPLC methods overcome the overestimation of bilirubin concentrations caused by the presence of unidentified diazo-positive compounds distinct from bilirubin. 40 High-performance liquid chromatography methods are also capable of differentiating various bilirubin isomers present under certain conditions in the biological systems. 41 Importantly, determination of the bilirubin subfractional changes, including detection of delta bilirubin, may help in the prediction of the risk of human diseases, such as cholestasis or gallstone disease, or in the differential diagnosis of such diseases. 42 , 43 Interestingly, the first method for separation and quantitative estimation of serum and biliary bilirubin fractions from serum and of three bilirubin fractions from bile was published as early as 1966. 44 Since that time, various chromatographic approaches were explored and a number of methods developed, including separation of native as well as derivatized bilirubins, such as ethyl anthranilate azo pigments or bilirubin methyl esters (for a review see ref. 44 ). These methods were gradually improved. The separation was enhanced by stepping from isocratic, normal-phase HPLC, to various gradient, reverse-phase systems. 45 The use of internal standards, such as xanthobilirubic acid 40 or mesobilirubin, 46 led to improvements in accuracy. Further enhancement was reached with the employment of mass spectrometric detection. 47 With these analytic advances, methods for the detection of bilirubin and its metabolites in tissues and cells were established. In contrast to the early insufficient attempts to determine bilirubin in brain tissue, which were based on spectrophotometry, 48 HPLC-based methods demonstrated much higher sensitivity and accuracy. Using a newly developed HPLC method based on C8-column separation with the implementation of the methanol/water/tetrabutyl ammonium hydroxide mobile phase and equipped with the diode array detector, it was possible to detect as little as 10 pmol of bilirubin per gram of tissue. 46 This method was used in experimental studies quantifying bilirubin in numerous organs, including the heart, 49 and especially brain tissues, 38 , 50 , 51 , 52 , 53 , 54 , 55 , 56 which are essential to understand the pathophysiology of bilirubin neurotoxicity.

Determination of bilirubin photoisomers and oxidation products

With increasing knowledge on the biological importance of bilirubin derivatives formed during oxidation processes, 57 sensitive and accurate analytic methods are becoming essential. These derivatives include tetra-, tri-, di-, and monopyrrolic bilirubin oxidation derivatives. Probably the most clinically important are bilirubin photoisomers formed during PT of severe unconjugated hyperbilirubinemia. However, the determination of these bilirubin derivatives in biological material is not trivial, because of the lack of commercial standards as well as instability of the pigments. In terms of the determination of bilirubin photoisomers, several methods were published. 58 Previously, an improved analytic HPLC method for the simultaneous determination of major bilirubin photoisomers, lumirubin, Z,E- and E,Z-bilirubins, and bilirubin was described using lumirubin as well as internal standards with tandem mass detection. 57 The method was validated on serum samples of jaundiced neonates treated with PT. It has the potential of facilitating our understanding of the kinetics and biology of bilirubin photoproducts, which to date are practically unknown. Research into other bilirubin oxidation products is also progressing. Tripyrrolic biopyrrins, which are clinically relevant markers of increased oxidative stress, can be analyzed by immunochemical methods using specific anti-bilirubin monoclonal antibodies. 59 Reliable analytic methods for dipyrrolic propentdyopents and monopyrrolic bilirubin oxidation products, Z-BOX A and B, with potential clinical implications were also published recently. 60 , 61 Finally, tetrapyrrolic compounds and their oxidation products were also studied in plants recently using these novel analytic methods and it will certainly improve our understanding of the biological relevance of these pigments. 62 , 63

Summary and conclusion

Invasive TSB measurements remain the gold standard on which the definitive diagnosis of SNH is based. According to international guidelines for neonatal jaundice management, the clinical decision for treatment of neonatal hyperbilirubinemia should be based on bilirubin levels measured in blood by diagnostic instrumentation. Any non-invasive bilirubin estimation must be confirmed by an invasive diagnostic method. Bench-top bilirubinometers and hand-held POC instruments have a few advantages over multiparameter instruments of being cheaper and faster. Test results are available immediately compared to results of TSB measurements using multiparameter instruments in a central laboratory. In addition, less blood is needed as the bench-top biliribinometers and hand-held POC instruments require minimal sample volumes. Novel POC bilirubin measurement methods, such as the BiliSpec and the Bilistick® System are of interest for many newborn infants, especially in LMICs, where the access to costly multiparameter instruments is limited. The main disadvantages of hand-held POC instruments and bench-top biliribinometers are that agreement with routine laboratory TSB varies and that they are still not included in EQA programs. TSB test results on these instruments should be accurate within permissible limits of measurement uncertainty to be fit for clinical purposes. The key to accomplish this is anchoring TSB test results to the latest internationally endorsed RMS for bilirubin. In addition, participation in EQA programs for TSB in the neonatal range, and close interdisciplinary cooperation between physicians and clinical chemists are needed to assure the desired analytic and clinical performance of TSB testing. It is surprising that after bilirubin was first measured two centuries ago, uncertainty still exists on how to correctly assess the concentration of this yellow pigment. Universal implementation of endorsed calibration hierarchies for test standardization remains a daunting task. Recently, analytic methods for bilirubin measurement in biological matrices, such as HPLC thermal lens spectrometry, spectrophotometric, molecular imprinting, and piezoelectric techniques were developed. These methods employ novel techniques that could further accelerate bilirubin research to improve the management of newborn infants with SNH.

Tiedemann, F. & Gmelin, L. In Die Verdauung nach Versuchen Ch. 10 (ed. Groos, K.) 79 (1826).

Frerichs, F. T. Klinik der Leberkrankheiten 1st edn, Vol. I of 2 volumes (1858).

Van den Bergh, A. A. H. & Snapper, J. Die Farbstoffe des Blutserums. Deut. Arch. Klin. Med. 110 , 540–561 (1913).

Google Scholar  

Malloy, H. T. & Evelyn, K. A. The determination of bilirubin with the photoelectric colorimeter. J. Biol. Chem. 119 , 481–490 (1937).

Article   CAS   Google Scholar  

Jendrassik, L. & Grof, P. Vereinfachte photometrische Methoden zur Bestimmung des Blutbilirubins. Biochem Z. 297 , 81–89 (1938).

CAS   Google Scholar  

BIPM, IEC, IFCC, ILAC, IUPAC, IUPAP, ISO, OIML. The International Vocabulary of Metrology—Basic And General Concepts and Associated Terms (VIM) 3rd edn. https://www.bipm.org/utils/common/documents/jcgm/JCGM_200_2012.pdf (2012).

Ngashangva, L., Bachu, V. & Goswami, P. Development of new methods for determination of bilirubin. J. Pharm. Biomed. Anal. 162 , 272–285 (2019).

Article   CAS   PubMed   Google Scholar  

Vreman, H. J. et al. Interlaboratory variability of bilirubin measurements. Clin. Chem. 42 , 869–873 (1996).

Doumas, B. T. & Eckfeldt, J. H. Errors in measurement of total bilirubin: a perennial problem. Clin. Chem. 42 , 845–848 (1996).

Cobbaert, C., Weykamp, C. & Hulzebos, C. V. Bilirubin standardization in the Netherlands: alignment within and between manufacturers. Clin. Chem. 56 , 872–873 (2010).

Greene, D. N., Liang, J., Holmes, D. T., Resch, A. & Lorey, T. S. Neonatal total bilirubin measurements: Still room for harmonization. Clin. Biochem. 47 , 1112–1115 (2014).

Kirk, J. M. Neonatal jaundice: a critical review of the role and practice of bilirubin analysis. Ann. Clin. Biochem. 45 , 452–462 (2008).

Lano, I. M., Lyon, A. W., Wang, L., Ruskin, R. & Lyon, M. E. Comparative evaluation of neonatal bilirubin using radiometer whole blood co-oximetry and plasma bilirubin methods from Roche diagnostics and ortho clinical diagnostics. Clin. Biochem. 53 , 88–92 (2018).

Wang, L. et al. Limitations and opportunities of whole blood bilirubin measurements by GEM premier 4000®. BMC Pediatr. 17 , 92 (2017).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Kazmierczak, S. C. et al. Direct spectrophotometric method for measurement of bilirubin in newborns: comparison with HPLC and an automated diazo method. Clin. Chem. 48 , 1096–1097 (2002).

Barko, H. A., Jackson, G. L. & Engle, W. D. Evaluation of a point-of-care direct spectrophotometric method for measurement of total serum bilirubin in term and near-term neonates. J. Perinatol. 26 , 100–105 (2006).

Lo, S. F., Jendrzejczak, B. & Doumas, B. T. Laboratory performance in neonatal bilirubin testing using commutable specimens: a progress report on a college of american pathologists study. Arch. Pathol. Lab Med. 132 , 1781–1785 (2008).

Article   PubMed   Google Scholar  

Olusanya, B. O., Ogunlesi, T. A. & Slusher, T. M. Why is kernicterus still a major cause of death and disability in low-income and middle-income countries? Arch. Dis. Child 99 , 1117–1121 (2014).

Slusher, T. M. et al. Burden of severe neonatal jaundice: a systematic review and meta-analysis. BMJ Paediatr. Open 1 , e000105 (2017).

Article   PubMed   PubMed Central   Google Scholar  

Keahey, P. A. et al. Point-of-care device to diagnose and monitor neonatal jaundice in low-resource settings. PNAS 114 , E10965–E10971 (2017).

Coda Zabetta, C. D. et al. Bilistick: a low-cost point-of-care system to measure total plasma bilirubin. Neonatology 103 , 177–181 (2013).

Greco, C. et al. Diagnostic performance analysis of the point-of-care Bilistick system in identifying SNH by a multi-country approach. EClinicalMedicine 1 , 14–20 (2018).

Greco, C. et al. Comparison between Bilistick System and transcutaneous bilirubin in assessing total bilirubin serum concentration in jaundiced newborns. J. Perinatol. 37 , 1028–1031 (2017).

Thielemans, L. et al. Laboratory validation and field usability assessment of a point-of-care test for serum bilirubin levels in neonates in a tropical setting. Version 2. Wellcome Open Res. 3 , 110 (2018).

Kamineni, B. et al. Accuracy of Bilistick (a point-of-care device) to detect neonatal hyperbilirubinemia. J. Trop. Pediatr. 66 , 630–636 (2020).

Rohsiswatmo, R. et al. Agreement test of transcutaneous bilirubin and bilistick with serum bilirubin in preterm infants receiving phototherapy. BMC Pediatr. 18 , 315 (2018).

Article   PubMed   PubMed Central   CAS   Google Scholar  

Boo, N.-Y. et al. The point-of-care Bilistick method has very short turn-around-time and high accuracy at lower cutoff levels to predict laboratory-measured TSB. Pediatr. Res. 86 , 216–220 (2019).

Article   PubMed   CAS   Google Scholar  

Tabatabaee, R. S., Golmohammadi, H. & Ahmadi, S. H. Easy diagnosis of jaundice: a smartphone-based nanosensor bioplatform using photoluminescent bacterial nanopaper for point-of-care diagnosis of hyperbilirubinemia. ACS Sens. 4 , 1063–1071 (2019).

Grohmann, K. et al. Bilirubin measurement for neonates: comparison of 9 frequently used methods. Pediatrics 117 , 1174–1183 (2006).

Lo, S. F. Laboratory accuracy in neonatal bilirubin: the search for truth in laboratory medicine. JAMA Pediatr. 170 , 529–530 (2016).

Stepman, H. C. M. et al. Measurements for 8 common analytes in native sera identify inadequate standardization among 6 routine laboratory assays. Clin. Chem. 60 , 855–863 (2014).

Vítek, L. Bilirubin as a predictor of diseases of civilization. Is it time to establish decision limits for serum bilirubin concentrations? Arch. Biochem. Biophys. 672 , 108062 (2019).

Jones, G. R. & Jackson, C. The Joint Committee for Traceability in Laboratory Medicine (JCTLM) - its history and operation. Clin. Chim. Acta 453 , 86–94 (2016).

Cobbaert, C., Smit, N. & Gillery, P. Metrological traceability and harmonization of medical tests: a quantum leap forward is needed to keep pace with globalization and stringent IVD-regulations in the 21st century! Clin. Chem. Lab Med. 56 , 1598–1602 (2018).

Klauke, R. et al. Reference measurement procedure for total bilirubin in serum re-evaluated and measurement uncertainty determined. Clin. Chim. Acta 481 , 115–120 (2018).

Fraser, C. G. & Peake, M. J. Problems associated with clinical chemistry quality control materials. CRC Crit. Rev. Clin. Lab Sci. 12 , 59–86 (1980).

Vitek, L. Bilirubin as a signaling molecule. Med. Res. Rev. 40 , 1335–1351 (2020).

Gazzin, S. et al. Bilirubin accumulation and Cyp mRNA expression in selected brain regions of jaundiced Gunn rat pups. Pediatr. Res. 71 , 653–660 (2012).

Adachi, Y. et al. Clinical application of serum bilirubin fractionation by simplified liquid chromatography. Clin. Chem. 34 , 385–388 (1988).

Muraca, M. & Blanckaert, N. Liquid-chromatographic assay and identification of mono- and diester conjugates of bilirubin in normal serum. Clin. Chem. 29 , 1767–1771 (1983).

Aziz, S. et al. Bilirubin-IX alpha and -IX beta pigments, coproporphyrins and bile acids in meconium and stools from full-term and preterm neonates during the first month of life. Acta Paediatr. 90 , 81–87 (2001).

Spivak, W. & Yuey, W. Application of a rapid and efficient h.p.l.c. method to measure bilirubin and its conjugates from native bile and in model bile systems. Potential use as a tool for kinetic reactions and as an aid in diagnosis of hepatobiliary disease. Biochem. J. 234 , 101–109 (1986).

Ostrea, E. M. Jr, Ongtengco, E. A., Tolia, V. A. & Apostol, E. The occurrence and significance of the bilirubin species, including delta bilirubin, in jaundiced infants. J. Pediatr. Gastroenterol. Nutr. 7 , 511–516 (1988).

Kuenzle, C. C., Sommerhalder, M., Ruttner, J. R. & Maier, C. Separation and quantitative estimation of four bilirubin fractions from serum and of three bilirubin fractions from bile. J. Lab Clin. Med. 67 , 282–293 (1966).

CAS   PubMed   Google Scholar  

Heirwegh, K. P., Fevery, J. & Blanckaert, N. Chromatographic analysis and structure determination of biliverdins and bilirubins. J. Chromatogr. 496 , 1–26 (1989).

Zelenka, J. et al. Highly sensitive method for quantitative determination of bilirubin in biological fluids and tissues. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 867 , 37–42 (2008).

Jašprova, J. et al. A novel accurate LC-MS/MS method for quantitative determination of Z-lumirubin. Sci. Rep. 10 , 4411 (2020).

Sawasaki, Y., Yamada, N. & Nakajima, H. Developmental features of cerebellar hypoplasia and brain bilirubin levels in a mutant (Gunn) rat with hereditary hyperbilirubinaemia. J. Neurochem. 27 , 577–583 (1976).

Muchova, L. et al. Statin treatment increases formation of carbon monoxide and bilirubin in mice: a novel mechanism of in vivo antioxidant protection. Can. J. Physiol. Pharm. 85 , 800–810 (2007).

Cuperus, F. J. et al. Beyond plasma bilirubin: the effects of phototherapy and albumin on brain bilirubin levels in Gunn rats. J. Hepatol. 58 , 134–140 (2013).

Schreuder, A. B. et al. Albumin administration protects against bilirubin-induced auditory brainstem dysfunction in Gunn rat pups. Liver Int. 33 , 1557–1565 (2013).

Schreuder, A. B. et al. Optimizing exchange transfusion for severe unconjugated hyperbilirubinemia: studies in the Gunn rat. PLoS ONE 8 , e77179 (2013).

Bortolussi, G. et al. Life-long correction of hyperbilirubinemia with a neonatal liver-specific AAV-mediated gene transfer in a lethal mouse model of Crigler-Najjar syndrome. Hum. Gene Ther. 25 , 844–855 (2014).

Vodret, S. et al. Albumin administration prevents neurological damage and death in a mouse model of SNH. Sci. Rep. 5 , 16203 (2015).

Vodret, S., Bortolussi, G., Jasprova, J., Vitek, L. & Muro, A. F. Inflammatory signature of cerebellar neurodegeneration during neonatal hyperbilirubinemia in Ugt1 -/- mouse model. J. Neuroinflam. 14 , 64 (2017).

Bockor, L. et al. Modulation of bilirubin neurotoxicity by the Abcb1 transporter in the Ugt1-/- lethal mouse model of neonatal hyperbilirubinemia. Hum. Mol. Genet. 26 , 145–157 (2017).

Jasprova, J. et al. The biological effects of bilirubin photoisomers. PLoS ONE 11 , e0148126 (2016).

Itoh, S., Isobe, K. & Onishi, S. Accurate and sensitive high-performance liquid chromatographic method for geometrical and structural photoisomers of bilirubin IX alpha using the relative molar absorptivity values. J. Chromatogr. A 848 , 169–177 (1999).

Vitek, L., Kraslova, I., Muchova, L., Novotny, L. & Yamaguchi, T. Urinary excretion of oxidative metabolites of bilirubin in subjects with Gilbert syndrome. J. Gastroenterol. Hepatol. 22 , 841–845 (2007).

Joerk, A. et al. Propentdyopents as heme degradation intermediates constrict mouse cerebral arterioles and are present in the cerebrospinal fluid of patients with subarachnoid hemorrhage. Circ. Res. 124 , e101–e114 (2019).

Clark, J. F., Loftspring, M., Wurster, W. L. & Pyne-Geithman, G. J. Chemical and biochemical oxidations in spinal fluid after subarachnoid hemorrhage. Front. Biosci. 13 , 1806–1812 (2008).

Konickova, R. Anti-cancer effects of blue-green alga Spirulina platensis, a natural source of bilirubin-like tetrapyrrolic compounds. Ann. Hepatol. 13 , 273–283 (2014).

Ritter, M. et al. Pyrrolic and dipyrrolic chlorophyll degradation products in plants and herbivores. Chemistry 26 , 6205–6213 (2020).

Download references

Acknowledgements

We greatly appreciate the help of T. van Wulfften Palthe in correcting the English grammar and language. The study was supported by grants NV18-07-00342 and RVO-VFN64165/2020 from the Czech Ministry of Health. The support of an intramural grant of Fondazione Italiana Fegato to Claudio Tiribelli is appreciated.

Author information

Authors and affiliations.

Beatrix Children’s Hospital, University Medical Center Groningen, Groningen, The Netherlands

Christian V. Hulzebos

Institute of Medical Biochemistry and Laboratory Diagnostics, and 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czech Republic

Libor Vitek & Aleš Dvořák

Bilimetrix s.r.l., Trieste, Italy

Carlos D. Coda Zabetta

Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, The Netherlands

Paul Schenk & Christa Cobbaert

Dutch Foundation for Quality Assessment in Medical Laboratories (SKML), Nijmegen, The Netherlands

Eline A. E. van der Hagen

MCA Laboratory, Queen Beatrix Hospital, Winterswijk, The Netherlands

Italian Liver Foundation, Trieste, Italy

Claudio Tiribelli

You can also search for this author in PubMed   Google Scholar

Contributions

Study concept and design, and retrieving and analyzing the literature: C.V.H., L.V., C.D.C.Z., C.C., and C.T. Drafting and critical revision of the manuscript for important intellectual content: C.V.H., L.V., C.D.C.Z., A.D., P.S., E.A.E.v.d.H., C.C., and C.T. All authors approved the final manuscript as submitted.

Corresponding author

Correspondence to Christian V. Hulzebos .

Ethics declarations

Competing interests.

Claudio Tiribelli is the President and Carlos D. Coda Zabetta is the CTO of Bilimetrix, the company responsible for the development of the Bilistick ® System. The remaining authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Hulzebos, C.V., Vitek, L., Coda Zabetta, C.D. et al. Diagnostic methods for neonatal hyperbilirubinemia: benefits, limitations, requirements, and novel developments. Pediatr Res 90 , 277–283 (2021). https://doi.org/10.1038/s41390-021-01546-y

Download citation

Received : 06 August 2020

Revised : 20 February 2021

Accepted : 10 April 2021

Published : 04 May 2021

Issue Date : August 2021

DOI : https://doi.org/10.1038/s41390-021-01546-y

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Using blue mini-leds as a light source designed a miniaturized optomechanical device for the detection of direct bilirubin.

  • Zhi Ting Ye
  • Hsin-Ching Kuo
  • Shang-Xuan Tsou

Nanoscale Research Letters (2022)

5 Tage altes Neugeborenes mit „Gelbsucht“

  • Norbert Teig

Monatsschrift Kinderheilkunde (2022)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

case study neonatal hyperbilirubinemia

This website is intended for healthcare professionals

British Journal Of Midwifery

  • { $refs.search.focus(); })" aria-controls="searchpanel" :aria-expanded="open" class="hidden lg:inline-flex justify-end text-gray-800 hover:text-primary py-2 px-4 lg:px-0 items-center text-base font-medium"> Search

Search menu

Abbas S, Nafea L, Abbas R Prevalence of ABO incompatibility and its effect on neonates' hyperbilirubinemia. Res J Pharm Technol.. 2020; 13 https://doi.org/10.5958/0974-360x.2020.00028.1

Abd Elmoktader A, Hussein S, Boraik M Hyperbilirubinemia in neonatal intensive care units: incidence and etiology at Fayoum University Hospital. Fayoum Univ Med J.. 2019; 3:8-14 https://doi.org/10.21608/fumj.2019.60476

Abu Mostafa S, Aljeesh Y, Abu Hamad K, Alnahhal M Risk factors of hyperbilirubinemia among admitted neonates in the Gaza Strip: case control study. Public Health Research. 2017; 7:39-45 https://doi.org/10.5923/j.phr.20170702.01

Acharya N, Paneru C Prevalence and etiology of neonatal jaundice in a tertiary care hospital. Journal of Nepalgunj Medical College. 2021; 18:(2)35-38 https://doi.org/10.3126/jngmc.v18i2.38891

Adoba P, Ephraim R, Kontor K Knowledge level and determinants of neonatal jaundice: a cross-sectional study in the Effutu Municipality of Ghana. Int J Pediatr.. 2018; 2018:1-9 https://doi.org/10.1155/2018/3901505

Aiswarya AT, Sajeeth CI The incidence, risk factors and management of Neonatal Jaundice in a Government hospital, Palakkad District, Kerala. Int J Health Sci Res.. 2016; 6:(6)123-129

Alkhaldi SM, Al-Kuran O, AlAdwan MM, Dabbah TA, Dalky HF, Badran E Determinants of breastfeeding attitudes of mothers in Jordan: a cross-sectional study. PLoS One. 2023; 18:(5) https://doi.org/10.1371/journal.pone.0285436

Al-Lawama M, Al-Rimawi E, Al-Shibi R, Badran E Adoption of the American Academy of Pediatrics' neonatal hyperbilirubinemia guidelines and its effect on blood exchange transfusion rate in a tertiary care center in Amman, Jordan. J Blood Med. 2018; 9:61-66 https://doi.org/10.2147/JBM.S162191

Awang H, Ja'afar SM, Ishak NAW, Dollah Z Determinants of neonatal jaundice among newborns in Pasir Puteh District, Kelantan. Int J Health Med Sci.. 2020; 6:109-122 https://doi.org/10.32827/ijphcs.6.6.109

Aynalem S, Abayneh M, Metaferia G Hyperbilirubinemia in preterm infants admitted to neonatal intensive care units in Ethiopia. Glob Pediatr Health. 2020; 7 https://doi.org/10.1177/2333794X20985809

Bhutani S, Klempel MC, Kroeger CM, Trepanowski JF, Varady KA Alternate day fasting and endurance exercise combine to reduce body weight and favorably alter plasma lipids in obese humans. Obesity. 2013; 21:(7)1370-1379 https://doi.org/10.1002/oby.20353

Bogale TM, Akanaw Bogale W, Guadie Kassie D, Woldesellassie A, Tamiru A Factors associated with neonatal hyperbilirubinemia in case files of all admitted inborn and outborn neonates in Northwest Ethiopia in 2019. Iran J Neonatol.. 2021; 12:46-53 https://doi.org/10.22038/ijn.2020.49279.1859

Boskabadi H, Rakhshanizadeh F, Zakerihamidi M Evaluation of maternal risk factors in neonatal hyperbilirubinemia. Arch Iran Med. 2020; 23:128-140

Bozkurt Ö, Yücesoy E, Oğuz B, Akinel Ö, Palali MF, Ataş N Severe neonatal hyperbilirubinemia in the southeast region of Turkey. Turk J Med Sci.. 2020; 50:(1)103-109 https://doi.org/10.3906/sag-1906-22

Brits H, Adendorff J, Huisamen D The prevalence of neonatal jaundice and risk factors in healthy term neonates at National District Hospital in Bloemfontein. Afr J Prim Health Care Fam Med. 2018; 10:1-6 https://doi.org/10.4102%2Fphcfm.v10i1.1582

Budi Wijaya A The relationship between mode of delivery and neonatal serum bilirubin in Melati Husada Women and Children Hospital, Malang. Paediatr Indones. 2017; 57

Calculator.net. Sample size calculator. 2024. https//www.calculator.net/sample-size-calculator.html?type=1&cl=95&ci=5&pp=33&ps=1000&x=Calculate (accessed 19 February 2024)

Devi D, Vijaykumar B Risk factors for neonatal hyperbilirubinemia: a case control study. Int J Reprod Contracept Obstet Gynecol.. 2017; 6:(1)198-202 https://doi.org/10.18203/2320-1770.ijrcog20164657

Faustina GA, Yunitasari E, Puspitasari D Risk factors neonatal jaundice – literature review. Nursing Update: Jurnal Ilmiah Ilmu Keperawatan. 2021; 12:(3)119-127 https://doi.org/10.36089/nu.v12i3.386

Haile MT Prevalence and contributing factors of neonatal jaundice in neonatal intensive care unit at St Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia, 2019. J Health Med Nurs. 2020; https://doi.org/10.7176/JHMN/74-03

Hassan B, Zakerihamidi M The correlation between frequency and duration of breastfeeding and the severity of neonatal hyperbilirubinemia. J Matern-Fetal Neonatal Med. 2017; 31:457-463 https://doi.org/10.1080/14767058.2017.1287897

Israel-Aina YT, Omoigberale AI Risk factors for neonatal jaundice in babies presenting at the University of Benin Teaching Hospital, Benin City. Niger J Paed. 2012; 39:(4)159-163 https://doi.org/10.4314/njp.v39i4.2

Jena MSS, Dash A Yellowness is a threat to newborn - a review. Asian J Pharm Clin Res.. 2018; 11:(2)

Karasneh RA, Migdady FH, Alzoubi KH, Al-Azzam SI, Khader YS, Nusair MB Trends in maternal characteristics, and maternal and neonatal outcomes of women with gestational diabetes: a study from Jordan. Ann Med Surg. 2021; 67 https://doi.org/10.1016/j.amsu.2021.102469

Khasawneh M, Rubaie Z, Khashashneh I, Makhlouf F, Alkafajei A Adherence with American Academy of Pediatrics guidelines when managing neonatal jaundice in Jordan. Res Reports Neonatol.. 2013; 3:27-31 https://doi.org/10.2147/RRN.S42868

Khasawneh W, Sindiani A, Rawabdeh SA, Aleshawi A, Kanaan D Indications and clinical profile of neonatal admissions: a cross-sectional descriptive analysis from a single academic center in Jordan. J Multidiscip Healthc.. 2020; 13:997-1006 https://doi.org/10.2147/jmdh.s275267

Lake E, Abera G, Azeze G Magnitude of neonatal jaundice and its associated factor in neonatal intensive care units of Mekelle City Public Hospitals, Northern Ethiopia. Drug Metab Dispos.. 2019; 1-9 https://doi.org/10.1155/2019/1054943

Li L, Wan W, Zhu C Breastfeeding after a cesarean section: a literature review. Midwifery. 2021; 103:103-117 https://doi.org/10.1016/j.midw.2021.103117

Mirghafourvand M, Seyedi R, Jannat Dost A, Mohammad-Alizadeh-Charandabi S, Asghari Jafarabadi M Relationship between neonatal skin bilirubin level and severe jaundice with maternal, childbirth, and neonatal characteristics. Iran J Neonatol.. 2019; 10:61-67 https://doi.org/10.22038/Ijn.2019.33282

Mojtahedi SY, Izadi A, Seirafi G, Khedmat L, Tavakolizadeh R Risk factors associated with neonatal jaundice: a cross-sectional study from Iran. Open Access Maced J Med Sci.. 2018; 6 https://doi.org/10.3889/oamjms.2018.319

Mullany LC, Katz J, Khatry SK, LeClerq SC, Darmstadt GL, Tielsch JM Neonatal hypothermia and associated risk factors among newborns of southern Nepal. BMC Med. 2010; 8 https://doi.org/10.1186/1741-7015-8-43

Murekatete C, Muteteli C, Nsengiyumva R, Chironda G Neonatal jaundice risk factors at a district hospital in Rwanda. RJMHS. 2020; 3:204-213 https://doi.org/10.4314/rjmhs.v3i2.10

Neal-Kluever A, Fisher J, Grylack L, Kakiuchi-Kiyota S, Halpern W Physiology of the neonatal gastrointestinal system relevant to the disposition of orally administered medications. Drug Metab Dispos.. 2018; 47:296-313 https://doi.org/10.1124/dmd.118.084418

Nyangabyaki-Twesigye C, Mworozi E, Namisi C Prevalence, factors associated and treatment outcome of hyperbilirubinaemia in neonates admitted to St Francis hospital, Nsambya, Uganda: a descriptive study. Afr Health Sci.. 2020; 20:(1)397-405 https://doi.org/10.4314/ahs.v20i1.46

Olatubi M, Ibitoye O, Sadibo O, Bolarinwa OS, Adamolekun MM Prevalence of neonatal jaundice at a tertiary health institution in Ondo state, Nigeria. J. Pre-Clin Clin Res.. 2019; 13:114-117 https://doi.org/10.26444/jpccr/111773

Onyearugha CN, Onyire BN, Ugboma HAA Neonatal jaundice: prevalence and associated factors as seen in Federal Medical Centre Abakaliki, Southeast Nigeria. Med Res.. 2011; 3:(3)40-45

Oppong J, Ampofo H, Boakye-Danquah C, Nsiah I Prevalence and risk factors associate with neonatal jaundice at Cape Coast Teaching Hospital (CCTH), Cape-Coast. Int J Innov Res Adv Stud. 2019; 6:(2)

Ricci S, Kyle T, Carman S Maternity and pediatric nursing.Netherlands: Wolters Kluwer; 2021

Sadeq TN, Feyad HF, Abdul Hameed GR Risk factors of neonatal jaundice at Al Kadhimiya Pediatrics Hospital in Baghdad, Iraq. JRUCS. 2019; 1:(44) https://doi.org/10.55562/jrucs.v44i1.139

Seyedi R, Mirghafourvand M, Jannat Dost A, Mohammad-Alizadeh-Charandabi S, Asghari Jafarabadi M Relationship between neonatal skin bilirubin level and severe jaundice with maternal, childbirth, and neonatal characteristics. IJN. 2019; 10:(2) https://doi.org/10.22038/ijn.2019.33282.1478

Slusher T, Zamora T, Appiah D Burden of severe neonatal jaundice: a systematic review and meta-analysis. BMJ Paediatr Open. 2017; 1 https://doi.org/10.1136/bmjpo-2017-000105

Shukla M, Agarwal M Knowledge of mothers regarding neonatal jaundice attending immunization clinic at a tertiary care hospital of Lucknow. Int J Appl Res.. 2016; 2:297-299

Tender KE Risk factors for sever hyperbilirubinemia (Jaundice) among neonates in Accra.Ghana: University of Ghana; 2018

West BA, Josiah AE Neonatal jaundice: prevalence, risk factors, and outcomes among inborn babies in a neonatal unit in Port Harcourt, Nigeria. IJBAMR. 2020; 9:92-103 https://doi.org/10.36848/IJBAMR/2020/18215.55572

Wongnate J Incidence and causes of neonatal jaundice in neonates at PhuKieochalermprakiat Hospital, Chaiyaphum Province. Chaiyaphum Med J.. 2021; 41:85-93

Yahya N, Yuniati T, Lubis L Characteristics of neonatal hyperbilirubinemia at West Java's Top Referral Hospital, Indonesia. Althea Med J.. 2017; 4:(2)167-172

Maternal and neonatal factors associated with neonatal jaundice in Jordan: a case-control study

Khitam Ibrahem Shlash Mohammad

Jordan University of Science & Technology, Jordan

View articles · Email Khitam Ibrahem Shlash

Maram Al–Shdefat

View articles

Suhaila Halasa

The University of Jordan, Jordan

Rachel Joseph

School of Nursing, Liberty University, USA

Mohammad Alafi

Medical student, Faculty of Medicine, Jordan University of Science and Technology, Jordan

Mohammed ALBashtawy

Community Health Nursing Department, Princess Salma Faculty of Nursing, Al Al-Bayt University, Jordan

Abdullah Alkhawaldeh

Asem Abdalrahim

Associate professor, Community Health Nursing Department, Princess Salma Faculty of Nursing, Al Al-Bayt University, Jordan

Malakeh Malak

Associate professor, Community Health Nursing Department, Faculty of Nursing, Al Zaetonah University, Jordan

Debra Creedy

Professor of Perinatal Mental Health, Maternal, Newborn and Families Research Collaborative, Menzies Institute of Health Queensland, Griffith University, Australia

Jenny Gamble

Professor and Head of Midwifery, Newborn and Families Research Collaborative, Menzies Institute of Health Queensland, Griffith University, Australia

case study neonatal hyperbilirubinemia

Background/Aims

Neonatal jaundice is a common cause of hospitalisation. This study aimed to identify maternal and neonatal factors associated with neonatal jaundice among hospitalised neonates in north Jordan.

This was a case control study involving 312 mothers and their neonates (106 cases, 206 controls), recruited from two governmental hospitals in north Jordan. A structured interview was used to collect data. Neonatal medical records were accessed for clinical data.

Maternal factors associated with higher risk of jaundice included having higher education, being employed and having had a caesarean section. Neonatal factors associated with higher risk of jaundice included being preterm, receiving <5 feeds per day and having ABO incompatibility.

Conclusions

Using evidence-based guidelines for the assessment and management of risk factors helps to reduce the prevalence of jaundice requiring hospitalisation among neonates. Raising mothers' awareness of neonatal jaundice through interventions and education sessions during antenatal care, in particular for high-risk mothers, is also likely to help reduce its prevalence.

Neonatal jaundice is a common cause of hospitalisation for both full-term and preterm neonates ( Mojtahedil et al, 2018 ). Almost all newborns have a total serum bilirubin level of >1mg/dL, the upper limit of normal for adults ( Aynalem et al, 2020 ). High levels (≥5mg/dL) manifest clinically as jaundice and, a consequence of increased breakdown of red blood cells and/or decreased hepatic excretion of bilirubin, producing a yellow-orange discoloration of the skin and sclera ( Brits et al, 2018 ; Olatubi et al, 2019 ). Neonatal jaundice commonly occurs in the first week after birth, although it is not harmful, is self-limiting and usually improves without treatment ( Awang et al, 2020 ; Ricci et al, 2021 ).

Factors associated with the development of neonatal jaundice include prematurity, ABO incompatibility, Rh incompatibility, glucose-6-phosphate dehydrogenase (G6PD), sepsis, breast problems (eg sore or cracked nipples or breast engorgement, associated with inadequate intake of breast milk), breastfeeding patterns, prolonged labor, having siblings who had neonatal jaundice, high birth weight, primiparity, male gender, mode of birth and maternal diabetes mellitus ( Olatubi et al, 2019 ; Seyedi et al, 2019 ; Abbas et al, 2020 ; Aynalem et al, 2020 ; Boskabadi et al, 2020 ; West and Josiah, 2020 ; Bogale et al, 2021 ; Karasneh et al, 2021 ; Wongnate, 2021 ). Acute bilirubin encephalopathy (kernicterus) is a major complications of hyperbilirubinemia, which can cause a spectrum of neurologic problems ( Aynalem et al, 2020 ). Surviving neonates may have long-term neurodevelopmental deficits, such as cerebral palsy, sensorineural hearing loss, intellectual difficulties and gross developmental delays ( Aynalem et al, 2020 ).

The prevalence of neonatal jaundice in high-income countries is low, affecting approximately 4.4 per 10 000 live births in America and 3.2 in Europe ( Slusher et al, 2017 ). In low- and middle-income countries, the prevalence is significantly higher; at 31.6% in Ethiopia ( Bogale et al, 2021 ), 35.9% in Nigeria ( Olatubi et al, 2019 ), 44.4% in Rwanda ( Murekatete et al, 2020 ), 55.2% in South Africa ( Brits et al, 2018 ) and 63% in South Asian countries, such as Malaysia ( Awang et al, 2020 ).

Little is known about the prevalence of neonatal jaundice in the Arab world. To the best of the authors' knowledge, only four published studies have been conducted on the prevalence of neonatal jaundice in the Middle Eastern region. These studies indicated higher prevalence rates than in high-income countries. The prevalence of neonatal jaundice in Iraq was 13.5%, with ABO incompatibility and Rh incompatibility identified as major risk factors (Sadeq et al, 2015). Another study from Iraq reported a 17.3% prevalence, and found that male neonates were at increased risk of developing neonatal jaundice than females ( Abbas et al, 2020 ). In Egypt the prevalence of neonatal jaundice was 45.6%, with ABO incompatibility, Rh incompatibility and G6PD found to be the leading causes of hyperbilirubinemia ( Abd Elmoktader et al, 2019 ). A case-control study in Gaza highlighted breastfeeding as a risk factor for the development of neonatal jaundice in comparison to bottlefeeding, but this may be because 74.7% of the case group were breastfeeding ( Abu Mostafa et al, 2017 ). Despite these findings, there is a lack of research on the association between maternal/neonatal factors and neonatal jaundice.

In Jordan, there has been no specific investigation of the prevalence and factors associated with the development of neonatal jaundice. There is also an absence of national guidelines for the evaluation and management of jaundice. Khasawneh et al (2013 ; 2020 ) determined that Jordanian doctors did not adhere to the American Academy of Pediatrics Guidelines (2004) when managing neonatal jaundice. More recently, an investigation of the clinical profile of neonatal admissions found that 10.7% were related to hyperbilirubinemia ( Khasawneh et al, 2020 ).

This study is intended to provide a baseline for determining the prevalence and correlated factors of neonatal jaundice, which can help healthcare providers develop appropriate management strategies. Therefore, the study aimed to investigate the prevalence of neonatal jaundice in Jordan, and the maternal and neonatal factors associated with its development.

A hospital-based unmatched case-control study design was used. A case-control design has many benefits, including that less time is required to conduct the study than is needed for a cohort study; this is because the condition has already occurred, which allows researchers to consider multiple risk factors simultaneously, and is useful in a preliminary study to establish an association.

For the purposes of this study, a case was defined as any neonate (term or preterm) admitted to the neonatal intensive care unit of one of the two selected hospitals and diagnosed as having jaundice by a pediatrician/physician, with a total serum bilirubin ≥5mg/dl as shown by laboratory test. A control was defined as any neonate (term or preterm) admitted to a neonatal intensive care unit with total serum bilirubin <5mg/dl and with no sign and symptoms of jaundice or liver disease. The two hospitals were purposively selected for the study, as they included neonatal intensive care units.

Based on a literature review, it was assumed that 33% of admissions to the neonatal intensive care units would have primary jaundice and there was a finite population size of 1000, according to the admission rates at the selected hospitals from 2020. The sample size was calculated using an online calculator ( Calculator.net, 2024 ) with a confidence level of 95% and a margin of error of 5%, yielding a sample size of 254 neonates. To account for non-response and incomplete records, the sample size was adjusted to approximately 300, to be split between 100 neonates admitted with jaundice as the primary cause (cases) and 200 neonates admitted for causes other than jaundice (controls).

As identifying matched control groups is time-consuming ( Shukla and Agarwal, 2016 ), convenience sampling was used. Consenting mothers who were Jordanian and could speak and read Arabic were included in the study. Neonates whose mothers did not consent or were diagnosed with liver problems were excluded from the study. Neonates with no laboratory proof of their total serum bilirubin level were also excluded.

Recruitment

Recruitment was conducted in the neonatal intensive care units of two government hospitals in the north of Jordan: the Princess Badeah Teaching Hospital and the Al-Mafraq Obstetrics and Gynecology Hospital. Confirmed cases of jaundice and control cases admitted between January and July 2021 were identified. Mothers were approached by a member of the team about the study when they came to visit their baby in the neonatal intensive care unit. Of the 810 mothers contacted, 362 met the inclusion criteria and were included in the study. A total of 50 mothers were excluded for missing data in medical records (n=13), withdrawal (n=22), and no response (n=15). The final sample of 312 consisted of 106 neonates who had developed jaundice (and their mothers, referred to as cases) and 206 neonates who had not developed jaundice (and their mothers, referred to as controls) giving a sample of 312 neonates (including both term and preterm neonates) and their mothers.

Data collection

Data were collected by reviewing the medical records of both neonates and their mothers. Identified mothers were interviewed using a structured questionnaire to validate the data from the medical records and collect any data not available in the record. The questionnaire was developed by the researchers based on the literature topic ( Mullany et al, 2010 ; Onyearugha et al, 2011 ; Israel-Aina and Omoigberale, 2012 ; Bhutani et al, 2013 ). The first section of the questionnaire gathered sought maternal sociodemographic characteristics. The second section collected neonatal data. The third and final section collected etiological factors.

The lead researcher met with the directors of participating hospitals and the chairperson of the neonatal intensive care unit to explain the purpose of the study and data collection. Participant mothers were interviewed by phone at a convenient time of their choice. All women were interviewed in their native language (Arabic) by research assistants who were trained in conducting interviews.

Data analysis

Data were analysed using the Statistical Package of Social Science (version 25). Frequencies means and standard deviations were calculated for demographic variables. Pearson's chi-square test was performed to determine associations between risk factors and neonatal jaundice. Multiple logistic regression analysis was performed to determine the odds ratios of risk factors that were significantly associated with neonatal jaundice. Multiple logistic regression analysis was performed to identify predictors of neonatal jaundice and establish the existence of specific differences among selected risk factors (independent factors). An alpha level of 0.05 was used for all statistical tests.

Ethical considerations

Ethical approval was obtained from the Institutional Review Board of Jordan University of Science and Technology (approval number: #724-2020) and from the Ministry of Health in Jordan. Mothers of eligible neonates who consented to participate were notified of confidentiality and their right to refuse participation, withdraw from the study at any time without explanation, not answer any questions and ask questions or clarifications at any time. Oral and written informed consent was obtained from the mothers. All communication was in Arabic.

Maternal demographic characteristics are shown in Table 1 . The largest proportion of participant mothers (38.5%) were 25–35 years old. Around one-third (35.9%) had completed high school, while 8.7% were illiterate. More than half were employed (54.5%). The largest proportion of mothers were in blood group A (28.8%), just over half (51.9%) had a vaginal birth and less than half (43.3%) had never had a baby who was jaundiced.

Characteristic Frequency, n (%)
Case, n=106 Control, n=206 Total, n=312
Age (years) <25 30 (28.3) 40 (19.4) 70 (22.4)
  25–35 42 (39.6) 78 (37.9) 120 (38.5)
  >35 28 (26.4) 84 (40.8) 112 (35.9)
  Missing 6 (5.7) 4 (1.9) 10 (3.2)
Education None 16 (15.1) 11 (5.3) 27 (8.7)
  Primary 30 (28.3) 41 (19.9) 71 (22.8)
  High school 45 (42.5) 67 (32.5) 112 (35.9)
  Bachelor or higher 15 (14.2) 83 (40.3) 98 (31.4)
  Missing 0 (0.0) 4 (1.9) 4 (1.3)
Occupation Employed 69 (65.1) 101 (49.0) 132 (42.3)
  Unemployed 31 (29.2) 101 (49.0) 170 (54.5)
  Missing 6 (5.7) 4 (1.9) 10 (3.2)
Blood group A 37 (34.9) 53(25.7) 90 (28.8)
  B 23 (21.7) 57(27.7) 80 (25.6)
  AB 22 (20.8) 48(23.3) 70 (22.4)
  O 24 (22.6) 48 (23.3) 72 (23.1)
Mode of birth Vaginal 71 (67.0) 91 (44.2) 162 (51.9)
Caesarean section 34 (32.1) 112 (54.4) 146 (46.8)
Assisted 1 1 Not included
Missing 1 (0.9) 3 (1.5) 4 (0.3)
History of baby with jaundice Yes 55 (51.9) 80 (38.8) 135 (43.3)
No 45 (42.5) 124 (60.2) 169 (54.2)
Missing 6 (5.7) 2 (1.0) 8 (2.5)

The demographic characteristics of the neonates are shown in Table 2 . More than half (57.4%) were preterm and the largest proportion (45.8%) had a birth weight of 2.5–4kg. Approximately half (53.8%) were female. One-third of neonates (30.8%) were in blood group B, more than half of the neonates (53.5%) were breastfeeding and nearly two-thirds (62.2%) were suckling properly. Mothers reported that 105 neonates (33.7%) had <5 feeds per day. A small proportion of neonates had possible ABO blood incompatibility (10.6%), Rhesus incompatibility (6.7%) or G6PD (4.8%).

Characteristic Frequency, n (%)
Case, n=106 Control, n=206 Total, n=312
Gestational age (weeks) <37 87 (82.1) 92 (44.7) 179 (57.4)
  ≥37 19 (17.9) 114 (55.3) 133 (42.6)
  Missing 0 (0.0) 0 (0.0) 0 (0.0)
Birth weight (kg) <2.5 43 (40.6) 45 (21.8) 88 (28.2)
  2.5–4.0 46 (43.4) 97 (47.1) 143 (45.8)
  <4.0 17 (16.0) 63 (30.6) 80 (25.6)
  Missing 0 (0.0) 1 (0.5) 1 (0.5)
Sex Male 61 (57.5) 83 (40.3) 144 (46.2)
  Female 45(42.5) 123 (59.7) 168 (53.8)
Blood group A 23 (21.7) 46 (22.3) 69 (22.1)
  B 24 (22.6) 72 (35.0) 96 (30.8)
  AB 25 (23.6) 50 (24.3) 75 (24.0)
  O 34 (32.1) 38 (18.4) 72 (23.1)
Feeding Breast 53 (50.0) 114 (55.3) 167 (53.5)
  Bottle 47 (44.3) 89 (43.2) 136 (43.6)
  Missing 6 (5.7) 3 (1.5) 9 (2.8)
Ability to suckle Yes 64 (64.4) 130 (63.1) 194 (62.1)
  No 36 (34.0) 74 (35.9) 110 (35.2)
  Missing 6 (5.7) 2 (1.0) 8 (2.5)
Feeds per day <5 48 (48.0) 57 (28.1) 105 (33.7)
  5–8 31 (31.0) 75 (36.9) 106 (34.0)
  9–12 21 (21.0) 71 (35.0) 92 (29.5)
  Missing 6 (1.93) 3 (9.66) 9 (2.9)
ABO blood incompatibility Yes 19 (18.1) 14 (6.8) 33 (10.6)
  No 86 (81.9) 192 (93.2) 278 (89.1)
  Missing 1 (0.3) 0 (0.0) 1 (0.3)
Rhesus disease Yes 9 (8.5) 12 (5.9) 21 (6.7)
  No 97 (91.5) 193 (94.1) 290 (92.9)
  Missing 0 (0.0) 1 (0.3) 1 (0.3)
G6PD Yes 7 (6.8) 8 (3.9) 15 (4.8)
  No 96 (93.2) 196 (96.1) 292 (96.6)
  Missing 3 (0.96) 2 (0.64) 5 (1.6)

Prevalence of neonatal jaundice

Of the 810 admissions to the neonatal intensive care unit during the data collection period (6 months), 170 (20.9%) were for jaundice. The time of onset varied, with 72.6% developing jaundice 24 hours after birth, and neonates had a mean total serum bilirubin of 18.06mg/dl (±3.81mg/dl). Less than a third (27.4%) developed jaundice within 24 hours of birth, with a mean total serum bilirubin of 14.44mg/dl (±4.00mg/dl). Almost half (49.1%) had severe jaundice, with a mean total serum bilirubin of 18.8mg/dl (±2.0mg/dl). Table 3 summarises the prevalence of different types and severity of jaundice among the cases.

Characteristic Frequency, n=106 (%) Mean total serum bilirubin ± standard deviation (mg/dl)
Time of onset (hours) Within 24 (pathological) 29 (27.4) 14.44 ± 4.00
After 24 (physiological) 77 (72.6) 18.06 ± 3.81
Severity Mild 3 (2.8) 8.6 ± 5.5
  Moderate 51 (48.1) 12.3 ± 2.8
  Severe 52 (49.1) 18.8 ± 2.0

Factors associated with neonatal jaundice

Pearson's chi-square test showed significant associations between total serum bilirubin and five maternal variables: age (P=0.03), education (P=0.001), occupation (P=0.002), mode of birth (P=0.001) and history of a baby with jaundice (P=0.009) ( Table 4 ). There were also significant associations with six neonatal variables: gestational age (P=0.001), birth weight(P=0.001), sex (P=0.004), blood group (P=0.028), number of daily feeds (P=0.002) and potential ABO incompatibility (P=0.002) ( Table 5 ).

Characteristic Frequency, n (%) X P value
Case Control
Age (years) <25 30 (28.3) 40 (19.4) 6.5 0.03
  25–35 42 (39.6) 78 (38.9)    
  >35 28 (26.4) 84 (40.8)    
  Missing 6 (5.7) 4 (1.9)    
Education None 16 (15.1) 11 (5.3) 26.81 0.001
  Primary 30 (28.3) 41 (19.9)    
  High school 45 (42.5) 67 (32.5)    
  Bachelor or higher 15 (14.2) 83 (40.3)    
  Missing 0 (0.0) 4 (1.9)    
Occupation Employed 69 (69.0) 101 (49.0) 9.8 0.002
  Unemployed 31 (31.0) 101 (49.0)    
  Missing 6 (5.7) 4 (1.9)    
Mode of birth Vaginal 71 (67.0) 91 (44.2) 14.4 0.001
  Caesarean 34 (32.1) 112 (54.4)    
  Missing 1 (0.9) 3 (1.5)    
History of baby with jaundice Yes 55 (51.9) 80 (38.8) 6.7 0.009
No 45 (42.5) 124 (60.2)    
Missing 6 (5.7) 2 (1.0)    
Blood group A 37 (34.9) 53 (25.7) 3.2 0.35
  B 23 (21.7) 57 (27.7)    
  AB 22 (20.8) 48 (23.3)    
  O 24 (22.6) 48 (23.3)    
  Missing 0 (0.0) 0 (0.0)    
Characteristic Frequency X P value
Case Control
Gestational age (weeks) <37 87 (82.1) 92 (44.7) 40.06 0.001
  ≥37 19 (17.9) 114 (55.3)    
Birth weight (kg) <2.5 43(40.6) 45 (21.8) 14.6 0.001
  2.5–4.0 46 (43.4) 97 (47.1)    
  >4.0 17 (16.0) 63 (30.6)    
  Missing 0 (0.0) 1 (0.5)    
Sex Male 61 (57.5) 83 (40.3) 8.3 0.004
  Female 45 (42.5) 123(59.7)    
Blood group A 23 (21.7) 46 (22.3) 9.1 0.028
  B 24 (22.6) 72 (35.0)    
  AB 25 (23.6) 50 (24.3)    
  O 34 (32.1) 38 (18.4)    
Feeds per day <5 48 (45.3) 57 (27.7) 12.6 0.002
  5–8 31 (29.2) 75 (36.4)    
  9–12 21 (19.8) 71 (34.5)    
  Missing 6 (5.7) 3 (1.5)    
ABO blood incompatibility Yes 19 (17.9) 14 (6.8) 9.3 0.002
  No 86 (81.1) 192 (93.2)    
  Missing 1 (0.9) 0 (0.0)    
Rhesus incompatibility Yes 9 (8.5) 12 (5.8) 0.77 0.380
  No 97 (91.5) 193 (93.7)    
  Missing 0 (0.0) 1 (0.5)    
Neonatal G6PD Yes 7 (6.6) 8 (3.9) 1.21 0.270
  No 96 (90.6) 196 (95.1)    
  Missing 3 (2.8) 2 (1.0)    

Maternal and neonatal factors associated with total serum bilirubin were entered into multiple logistic regression ( Table 6 ). Of the maternal factors, only education, occupation and mode of birth were significantly associated with jaundice. Lower education (P=0.001), being employed (P=0.001) and having a caesarean section (P=0.003) increased the odds of developing jaundice. Among the neonatal factors, gestational age, number of feeds per day and ABO incompatibility were significantly related to jaundice. Preterm neonates (P=0.02), those who had fewer feeds per day (P<0.001) and those who were ABO incompatible (P=0.01) were more likely to develop jaundice.

Variable Subgroup odds ratio P value Crude odds ratio (95% confidence interval) Adjusted odds ratio (95% confidence interval) P value
Education None Reference        
  Primary 0.30 (0.08–1.08) 0.060 0.82 (0.63–1.07) 0.47 (0.34–0.66) 0.001
  Secondary 0.30 (0.90–1.00) 0.050      
  High school 0.04 (0.01–0.17) 0.001      
Occupation Employed Reference        
  Unemployed 0.32 (0.15–0.64) 0.001 0.67 (0.49–0.90) 0.55 (0.32–0.93) 0.001
Mode of birth Vaginal Reference        
  Caesarean 0.24 (0.12–0.49) 0.001 0.90 (0.73–1.12) 1.01 (0.94–1.09) 0.003
Gestational age (weeks) <37 Reference        
  ≥37 0.14 (0.06–0.30) 0.008 1.2 (1.01–1.58) 0.19 (0.10–0.38) 0.020
Feeds per day <5 Reference        
  5–8 0.42 (0.10–1.77) 0.03 0.24 (0.17–0.35) 0.72 (0.50–1.03) <0.001
  9–12 0.20 (0.04–0.87) 0.01      
ABO incompatibility No Reference        
  Yes 3.48 (1.21–9.98) 0.059 2.1 (1.32–3.46) 3.59 (1.35–9.58) 0.010

This mismatched case-control study to examine factors associated with neonatal jaundice is the first targeted study of neonatal jaundice in Jordan, to the authors' knowledge. The prevalence of neonatal jaundice was 20.9%, which is consistent with rates reported in China (20.3%) and Uganda (22.7%) (Awang et al, 2019; Nyangabyaki-Twesigye et al, 2020 ). The prevalence was lower than some other low- and middle-income countries, such as Africa, Nepal, India, South Asia and Egypt, where the prevalence of neonatal jaundice is repotedly between 31% and 67% ( Brits et al, 2018 ; Abd Elmoktader et al, 2019 ; Awang et al, 2019; Olatubi et al, 2019 ; Oppong et al, 2019 ; Bogale et al, 2021 ). Lower rates have been reported in Iraq (13.5%) ( Sadeq et al, 2019 ), Ethiopia (13.3%) ( Haile, 2020 ), Indonesia (4.1%) ( Yahya et al, 2017 ) and Turkey (3.6%) (Bozkurt et al, 2019). These differences may be the result of variations in study design, setting, time and method of data collection. However, they may also be the result of differences in socio-cultural and economic conditions, level of obstetrics care, and gestational age ( Tender, 2018 ).

Several demographic and birth-related variables were associated with the development of neonatal jaundice. Neonates of educated mothers were at higher risk of developing jaundice than babies of less educated mothers. This is consistent with previous studies, which have reported that neonates of educated mothers are at increased risk of jaundice ( Aiswarya and Sajeeth, 2016 ). Women with higher education are more likely to be employed, which was also significantly linked with increased risk of jaundice in both the present study and in the wider literature ( Aiswarya and Sajeeth, 2016 ; Adoba et al, 2018 ; Tender, 2018 ).

Higher education and employment can impact infant feeding, and number of feeds was significantly associated with neonatal jaundice both in the present study and in the literature ( Adoba et al, 2018 ; Alkhaldi et al, 2023 ). These women may prefer an infant feeding method and schedule that will not restrict their daily activities ( Alkhaldi et al, 2023 ). This may be linked to a preference for formula feeding, although feeding method was not significantly associated with jaundice. However, breastfeeding promotion programmes and initiatives that educate women on the role of breastfeeding may be beneficial.

Mode of birth was linked with the development of neonatal jaundice, with caesarean section increasing the risk of jaundice. This is similar to studies in Malaysia and South Africa, where babies born via a caesarean section were at increased risk of jaundice than those born via vaginal birth ( Budi Wijaya, 2017 ; Awang et al, 2019; Murekatete et al, 2020 ). This may be related to the anesthesia used during caesarean section (isoflurane and bupivacaine), which could increase bilirubin levels in neonates ( Budi Wijaya, 2017 ). In addition, mothers who gave birth via caesarean section are more likely to experience breastfeeding problems ( Li et al, 2021 ), which, as explained above, can be linked with higher risk of jaundice.

In the present study, prematurity was significantly associated with the development of neonatal jaundice. A preterm neonate (born before 37 weeks' gestation) may not be capable of eliminating bilirubin as rapidly as full-term babies, who have a more mature liver ( Jena and Dash, 2018 ). This is consistent with most previous studies, which emphasised the risks associated with prematurity ( Devi and Vijaykumar, 2017 ; Oppong et al, 2019 ; Boskabadi et al, 2020 ; Murekatete et al, 2020 ; West and Josiah, 2020 ; Acharya and Paneru, 2021 ; Wongnate, 2021 ). Although one study from Ghana reported that prematurity was not a significant factor in the development of neonatal jaundice, only a small proportion of their sample were preterm babies ( Tender, 2018 ).

While it is known that ABO incompatibility can result in neonatal jaundice ( Abd Elmoktader et al, 2019 ; Lake et al, 2019 ; Sadeq et al, 2019 ; Boskabadi et al, 2020 ; Murekatete et al, 2020 ; West and Josiah, 2020 ; Bogale et al, 2021 ; Faustina et al, 2021 ; Wongnate, 2021 ), the findings from this study demonstrated that neonates with ABO blood incompatibility were over three times as likely to develop neonatal jaundice. ABO incompatibility leads to hemolysis disorder (destruction of red blood cells), increasing the level of bilirubin in the blood ( Ricci et al, 2021 ).

Fewer daily feeds increased the odds of developing jaundice. This is may be because a reduction in the volume and frequency of feeding can hinder defecation and the removal of bilirubin through the bowels ( Neal-Kluever et al, 2018 ). Similarly, frequent feeding prevents dehydration and facilitates frequent urination ( Neal-Kluever et al, 2018 ). Several other studies have also reported that decreased frequency of feeding and feeding problems increase the risk for neonatal jaundice ( Hassan and Zakerihamidi, 2017 ; Seyedi et al, 2019 ; Haile, 2020 ).

Limitations

This study had several limitations, including using a mismatched case-control design and selection bias, which limited the researchers' ability to identify the temporal relationship between various risk factors and neonatal jaundice. In addition, this study was conducted in one region, and more studies are needed in different settings to confirm the findings.

Implications for practice

To improve neonatal survival and reduce severe morbidities, early identification of risk factors for jaundice is vital, so that appropriate interventions can be implemented. It is essential to integrate accurate assessment as part of routine care for mother and neonate, in order to decrease the prevalence of this disorder, which is considered a neglected element of clinical practice in Jordan ( Al-Lawama et al, 2018 ). Healthcare providers need education and training on the prevalence, risk factors and negative effects of jaundice for mothers, infants and families as a whole.

Raising awareness among mothers by conducting antenatal education is recommended, particularly for high-risk mothers. In Jordan, there is no national practice guideline for the assessment and management of neonatal jaundice. Decision-makers should work to establish a national guideline to screen high-risk neonates for jaundice and follow up with them during the postnatal period. Using evidence-based guidelines for the assessment and management of risk factors of jaundice helps to reduce the prevalence of jaundice among neonates ( Al-Lawama et al, 2018 ). Addressing neonatal jaundice is also likely to reduce length of stay in intensive care, additionally reducing financial strains on the family and healthcare institutions.

The present study showed significant associations between a mother's education and occupation, mode of childbirth, prematurity, ABO incompatibility, reduced feeding, and the development of jaundice among neonates in northern Jordan. Identifying these factors will allow neonatal nurses to educate parents in an effort to prevent jaundice. Further research on management of jaundice and development of national guidelines may also allow healthcare providers address the issue. Preventing prematurity and low birth weight in babies also will be beneficial.

  • Neonatal jaundice is a common cause of neonatal morbidity.
  • Identifying at-risk factors will allow prevention and lessen length of hospital stay for the neonates.
  • It is essential to integrate accurate assessment as a part of routine care provided to the mother and her neonate to decrease the prevalence of this disorder.
  • Using evidence-based guidelines for the assessment and management of risk factors of jaundice helps in reducing the prevalence of jaundice among neonates.

Case Study: Neonatal Jaundice


Neonatal Jaundice

Case Presentation

Martin and Kim were both twenty-five when they had Michael, their first child. Kim remained very healthy during her pregnancy and went into labor at 9:00 a.m., just 3 days after her due date. Delivery went quite smoothly, and that evening, mother and child rested comfortably. Two days later, Kim and Michael were released from the hospital. That evening at feeding time, Kim noticed that the whites of Michael's eyes seemed just slightly yellow, a condition that worsened noticeably by the next morning. Kim called the pediatrician and made an appointment for that morning.

Upon examining Michael, the pediatrician informed Martin and Kim that the infant had neonatal jaundice, a condition quite common in newborns and one that need not cause them too much concern. The physician explained that neonatal jaundice was the result of the normal destruction of old or worn fetal red blood cells and the inability of the newborn's liver to effectively process bilirubin, a chemical produced when red blood cells are destroyed. The physician told the parents he would like to see Michael every other day in order to monitor blood bilirubin concentration until the bilirubin concentration dropped into the normal range. He recommended that Kim feed Michael frequently and instructed them to place Michael in sunlight whenever possible.

Case Background

Neonatal jaundice in a disorder that affects nearly 50% of all newborns to at least a small degree. The yellow coloration of the skin and sclera of the eyes is due to the accumulation of bilirubin in adipose tissue and its adherence to collagen fibers. In neonatal jaundice, the excess bilirubin is not due to an abnormal level of red blood cell destruction. It is due to the inability of the young liver cells to conjugate bilirubin, or make it soluble in bile, so that it can be excreted and removed from the body by the digestive tract. This inability is corrected, usually within one week, as the liver cells synthesize the conjugation enzymes. If uncorrected, sufficiently high bilirubin concentrations can cause brain damage. Frequent feedings of a newborn with jaundice increase gastrointestinal tract motility and decrease the likelihood of reabsorbing significant amounts of bilirubin in the small intestine. Radiation from sunlight alters the chemical form of bilirubin, making is easier for the liver to excrete.

1
2
3
Date:
My name:
Section ID:
E-mail address:Format:
Me:
My Instructor:
My TA:
Other:
To learn more about the book this website supports, please visit its .
Copyright
Any use is subject to the and |
You must be a registered user to view the in this website.

If you already have a username and password, enter it below. If your textbook came with a card and this is your first visit to this site, you can to register.
Username:
Password:
'); document.write(''); } // -->
( )
.'); } else{ document.write('This form changes settings for this website only.'); } //-->
Send mail as:
'); } else { document.write(' '); } } else { document.write(' '); } // -->
'); } else { document.write(' '); } } else { document.write(' '); } document.write('
TA email: '); } else { document.write(' '); } } else { document.write(' '); } // -->
Other email: '); } else { document.write(' '); } } else { document.write(' '); } // -->
"Floating" navigation? '); } else if (floatNav == 2) { document.write(' '); } else { document.write(' '); } // -->
Drawer speed: '; theseOptions += (glideSpeed == 1) ? ' ' : ' ' ; theseOptions += (glideSpeed == 2) ? ' ' : ' ' ; theseOptions += (glideSpeed == 3) ? ' ' : ' ' ; theseOptions += (glideSpeed == 4) ? ' ' : ' ' ; theseOptions += (glideSpeed == 5) ? ' ' : ' ' ; theseOptions += (glideSpeed == 6) ? ' ' : ' ' ; document.write(theseOptions); // -->
1. (optional) Enter a note here:

2. (optional) Select some text on the page (or do this before you open the "Notes" drawer).
3.Highlighter Color:
4.
Search for:
Search in:
Course-wide Content






News, Articles & Links

Quizzes












More Resources





Instructor Resources





Course-wide Content






4 Hyperbilirubinemia (Jaundice) Nursing Care Plans

Jaundice Nursing Care Plans and Nursing Diagnosis

Hyperbilirubinemia is the elevation of serum bilirubin levels that is related to the hemolysis of RBCs and subsequent reabsorption of unconjugated bilirubin from the small intestines. The condition may be benign or place the neonate at risk for multiple complications/untoward effects.

The newborn ‘s liver is immature, which contributes to icterus, or jaundice. The liver cannot clear the blood of bile pigments that result from the normal postnatal destruction of red blood cells. The higher the blood bilirubin level is, the deeper jaundice and the greater risk for neurological damage. Physiological jaundice is normal, while pathological jaundice is more serious, which occurs within 24 hours of birth, and is secondary to an abnormal condition, such ABO- Rh incompatibility . The normal rise in bilirubin levels in preterm infants is slower than in full-term infants. It lasts longer, which predisposes the infant to hyperbilirubinemia or excessive bilirubin levels in the blood.

Physiological jaundice is the most common type of newborn hyperbilirubinemia. This unconjugated hyperbilirubinemia presents in newborns after 24 hours of life and can last up to the first week. Pathological jaundice is defined as the appearance of jaundice in the first 24 hours of life due to an increase in serum bilirubin levels greater than 5 mg/dl/day, conjugated bilirubin levels ≥ 20% of total serum bilirubin, peak levels higher than the normal range, and the presence of clinical jaundice greater than two weeks. Breast milk jaundice occurs in breastfed newborns between the first and third day of life but peaks by day 5 to 15, with a decline occurring by the third week of life (Morrison, 2021).

In the past, hemolytic disease of the newborn was most often caused by an Rh blood type incompatibility. Because the prevention of Rh antibody formation has been available for almost 50 years, the disorder is now most often caused by an ABO incompatibility. In both instances, because the fetus has a different blood type than the mother, the mother builds antibodies against the fetal red blood cells, leading to hemolysis of the cells, severe anemia , and hyperbilirubinemia.

Table of Contents

Nursing problem priorities, nursing assessment, nursing diagnosis, nursing goals, 1. initiating patient education and health teachings, 2. promoting safety and preventing injuries and complications, 3. administer medications and provide pharmacologic support, 4. monitoring results of diagnostic and laboratory procedures, recommended resources, references and resources, nursing care plans and management.

The nursing care plan for clients with hyperbilirubinemia involves preventing injury /progression of the condition, providing support/appropriate information to family, maintaining physiological homeostasis with bilirubin levels declining, and preventing complications.

The following are the nursing priorities for patients with hyperbilirubinemia (jaundice):

  • Bilirubin level monitoring. Regularly monitoring the bilirubin levels in the patient’s blood to assess the severity of hyperbilirubinemia.
  • Identification of underlying cause. Investigating and identifying the underlying cause of hyperbilirubinemia to guide treatment decisions.
  • Phototherapy. Initiating and managing phototherapy to help break down bilirubin and reduce its levels in the blood.
  • Blood transfusion . Considering blood transfusion in severe cases of hyperbilirubinemia to remove excess bilirubin and provide additional red blood cells.
  • Neonatal assessment . Conducting a thorough neonatal assessment to evaluate the overall health and identify any additional concerns associated with hyperbilirubinemia.
  • Parent education. Educating parents about the causes, management, and signs of worsening hyperbilirubinemia, as well as the importance of follow-up care.
  • Liver function evaluation . Assessing liver function to determine if there are any underlying liver disorders contributing to hyperbilirubinemia.
  • Coordinating with pediatric specialists. Collaborating with pediatricians and specialists to ensure comprehensive care and appropriate management of hyperbilirubinemia.
  • Support for breastfeeding . Providing guidance and support to breastfeeding mothers to optimize feeding practices, which can help with bilirubin elimination.
  • Long-term follow-up. Planning for long-term follow-up to monitor the resolution of hyperbilirubinemia and identify any potential long-term effects or complications.

Assess for the following subjective and objective data :

  • See nursing assessment cues under Nursing Interventions and Actions.

Following a thorough assessment, a nursing diagnosis is formulated to specifically address the challenges associated with hyperbilirubinemia (jaundice) based on the nurse ’s clinical judgment and understanding of the patient’s unique health condition. While nursing diagnoses serve as a framework for organizing care, their usefulness may vary in different clinical situations. In real-life clinical settings, it is important to note that the use of specific nursing diagnostic labels may not be as prominent or commonly utilized as other components of the care plan. It is ultimately the nurse’s clinical expertise and judgment that shape the care plan to meet the unique needs of each patient, prioritizing their health concerns and priorities.

Goals and expected outcomes may include:

  • The mother will verbalize understanding of the cause, treatment, and possible outcomes of hyperbilirubinemia.
  • The mother will identify signs/symptoms requiring prompt notification of the healthcare provider.
  • The mother will demonstrate appropriate care for the infant.
  • The neonate will display indirect bilirubin levels below 12 mg/dl in term infants at three days of age.
  • The neonate will show resolution of jaundice by the end of the 1st wk of life.
  • The neonate will be free of CNS involvement.
  • The neonate will complete the exchange transfusion without complications.
  • The neonate will display decreasing serum bilirubin levels.
  • The neonate will maintain body temperature and fluid balance within the normal limits.
  • The neonate will be free of skin/tissue injury.
  • The neonate will demonstrate expected interaction patterns.

Nursing Interventions and Actions

Therapeutic interventions and nursing actions for patients with hyperbilirubinemia (jaundice) may include:

Neonatal jaundice is the main reason for admission from home to a neonatal unit. Many neonates are readmitted with extreme hyperbilirubinemia or bilirubin encephalopathy at or around day five and had been discharged as healthy from birth hospitalization. As the newborn is usually at home at the time of the bilirubin peaking, much of the onus for detecting the development of severe hyperbilirubinemia and evaluating the success of breastfeeding falls on the parents and community medical services (Kaplan et al., 2019).

Assess the family situation and support systems. Parents need guidance throughout the infant’s hospitalization to help to prepare them for this new experience. The mother is usually concerned with her ability to care for such a small and helpless creature. When she feels ready, she may assist the nurse in diapering, bathing , feeding , and other activities. Often the mother is discharged without her infant. This is difficult for the entire family and complicates attachment and bonding.

Assess the client’s and family members ’ knowledge and level of understanding. This helps in determining specific needs and clarifying previous information. The client and her family are assessed for their understanding of the diagnosis and their ability to cope during the unexpected extended period of recovery.

Provide parents with an appropriate written explanation of home phototherapy, listing technique and potential problems, and safety precautions. Non-specific written instructions are most likely a key factor contributing to the low attendance rate for early community follow-up for jaundice, as studied by Kaplan et al.. Some mothers provided reasons contributing to poor attendance. Poor understanding and insufficient explanation of the potential dangers of hyperbilirubinemia were leading factors. It is possible that the medical/nursing team, at the time of discharge, did not fully expound to parents the full reasons necessitating early follow-up (Kaplan et al., 2019).

Discuss appropriate monitoring of home therapy, e.g., periodic recording of infant’s weight, feedings, intake/output, stools, temperature, and proper reporting of infant status. Home phototherapy is recommended only for full-term infants after the first 48 hr of life, whose serum bilirubin levels are between 14 and 18 mg/dl with no increase in direct reacting bilirubin concentration. Nowadays, home phototherapy is very popular due to the importance of preventing mother-infant separation and continuity of care at home (Morrison, 2021).

Provide information about the types of jaundice, pathophysiological factors, and future implications of hyperbilirubinemia. Encourage to ask questions; reinforce or clarify information as needed. This promotes understanding the disease condition, correction of misconceptions, and reducing feelings of guilt and fear . Neonatal jaundice may be pathological, physiological, or breast milk–induced in etiology. Parents need an explanation of the rationale for phototherapy and why their infant needs it. Although phototherapy has not been used long enough that long-term effects can be studied, there appears to be minimal risk to an infant from the procedure, provided the infant’s eyes remain covered, and dehydration from increased insensitive water loss does not occur.

Discuss home management of mild or moderate physiological jaundice, including increased feedings, diffused exposure to sunlight (checking infant frequently), and a follow-up serum testing program. Parents’ understanding helps foster their cooperation once the infant is discharged. The information helps parents carry out home management safely and appropriately and recognize the importance of all aspects of the management program. Note: Exposure to direct sunlight is contraindicated as an infant’s tender skin is highly susceptible to thermal injury. Even though there is no evidence so far that infants who received phototherapy are at greater risk for developing skin cancer , all infants who receive phototherapy should (as should all infants) have sunscreen applied when they are in the sun and follow-up assessments in the coming years to detect skin cancer that possibly could occur from the therapy.

Provide information about maintaining milk supply through a breast pump and reinstating breastfeeding when jaundice necessitates interruption of breastfeeding. This helps mothers maintain adequate milk supply to meet the infant’s needs when breastfeeding is resumed. Infants weighing more than 1500 g (3.3 lb) may be able to bottle feed if a small, soft nipple with a large hole is used to minimize the energy and effort required for sucking. Breast milk may be manually expressed by the mother and placed in a bottle for her preterm infant.

Demonstrate means of assessing the infant for increasing bilirubin levels (e.g., blanching the skin with digital pressure to reveal the color of the skin, weight monitoring, or behavioral changes), especially if the infant is to be discharged early. To aid the parents in recognizing signs and symptoms of increasing bilirubin levels. Observing the infant’s skin, sclera , and mucous membranes for jaundice is included in the nursing care. Blanching of the skin over bony prominences enhances the evaluation for jaundice. Observing and reporting the progression of jaundice from the face to the abdomen and feet is important because the progression may indicate increasing bilirubin levels.

Provide parents with a 24-hr emergency telephone number and the name of the contact person, stressing the importance of reporting increased jaundice This decreases anxiety and prepares an immediate seek timely medical evaluation /intervention. Increased awareness of the importance of jaundice and early referring to hospitals among families can help reduce the complications of jaundice (Sardari et al., 2019).

Review rationale for specific hospital procedures/therapeutic interventions (e.g., phototherapy, exchange transfusions) and changes in bilirubin levels, especially if the neonate must remain in the hospital for treatment while the mother is discharged. This assists parents in understanding the importance of therapy, keep parents informed about the infant’s status and promotes informed decision-making . Note: Some hospitals have overnight rooms that allow the mother/father to remain with the infant. The use of intensive phototherapy in conjunction with hydration and close monitoring of serum bilirubin levels has greatly reduced the need for exchange transfusions. Exchange transfusion reduces the serum concentration of indirect bilirubin and can prevent heart failure in infants with severe anemia or polycythemia.

Discuss possible long-term effects of hyperbilirubinemia and the need for continued assessment and early intervention. Kernicterus is caused by a high bilirubin level in a baby’s blood. If left untreated, the bilirubin can then spread into the brain , where it causes long-term damage, which includes cerebral palsy , mental retardation, sensory difficulties, delayed speech, poor muscle coordination , learning difficulties, and enamel hypoplasia or yellowish-green staining of teeth, and even death .

Discuss the need for Rh immune globulin (RhIg) within 72 hours following delivery for an Rh-negative mother with an Rh-positive infant who has not been previously sensitized. Rh-Ig may minimize the incidence of maternal isoimmunization in non-sensitized mothers and may help to prevent erythroblastosis fetalis in subsequent pregnancies. Rh incompatibility is not commonly seen today because if Rh-negative women receive Rho immune globulin (RHIG or RhoGAM) within 72 hours after the birth of an Rh-positive newborn, the process of antibody formation will be halted, and sensitization will not occur.

Make appropriate arrangements for follow-up serum bilirubin testing at the same laboratory facility. Treatment is discontinued once serum bilirubin concentrations fall below 14 mg/dl, but serum levels must be rechecked in 12–24 hr to detect possible rebound hyperbilirubinemia. Although phototherapy may prevent an increase in bilirubin levels, it does not affect the underlying cause of jaundice. If phototherapy fails to keep the total serum bilirubin at acceptable levels to prevent kernicterus, an exchange transfusion may be indicated.

Provide appropriate referral for a home phototherapy program, if necessary. The lack of available support systems and education may necessitate visiting nurses to monitor the home phototherapy program. /Home phototherapy programs are being used for newborns with mild to moderate physiological jaundice. The infant’s pediatrician makes a referral for home care based on the newborn’s health, bilirubin levels (generally between 10 to 14 mg/dL), evidence of jaundice, and the family’s suitability for complying with the home program.

Educate the parents regarding home phototherapy. The parents can use a phototherapy blanket in a bassinet or a fiberoptic pad for home phototherapy. These allow the infant to be held, reducing the risk of eye damage. Written instructions are given to parents. Parents keep a daily record of their infant’s temperature, weight, intake and output , stools, and feedings. The parents must ensure that the infant’s eyes are covered under the lights to prevent injury to the infant’s retina and place a small diaper over the infant’s gonad area to protect their ovaries or testes.

Promoting safety and preventing injuries and complications in patients with hyperbilirubinemia (jaundice) involves implementing measures to mitigate the risks associated with elevated bilirubin levels. This includes strict adherence to phototherapy protocols, ensuring proper eye protection during phototherapy sessions, monitoring vital signs and hydration status, closely observing for signs of neurotoxicity or kernicterus, promptly addressing any concerning symptoms, providing education to parents on safe handling and care practices, and maintaining a collaborative approach among healthcare professionals to promptly identify and manage any complications that may arise.

Assess infant/maternal blood group and blood type. ABO incompatibility affects 20% of all pregnancies and most commonly occurs in mothers with type O blood, whose anti-A and anti-B antibodies pass into fetal circulation, causing RBC agglutination and hemolysis. ABO and Rh incompatibilities increase the risk for jaundice. Maternal antibodies cross the placenta in Rh-negative women who had previously been sensitized due to Rh-positive infants. Antibodies attach to fetal RBCs and increase the risk of hemolysis.

Assess the infant in daylight. This prevents distortion of actual skin color through the use of artificial lighting. Most infants do not appear jaundiced at birth because the maternal circulation has evacuated the rising indirect bilirubin level. With birth, progressive jaundice, usually occurring within the first 24 hours of life, will begin, indicating that a hemolytic process is occurring in both Rh and ABO incompatibility.

Review infant’s condition at birth, noting the need for resuscitation or evidence of excessive ecchymosis or petechiae, cold stress, asphyxia, or acidosis. Asphyxia and acidosis reduce the affinity of bilirubin to albumin. A study found that perinatal asphyxia was negatively associated with neonatal hyperbilirubinemia. This might be explained by acidosis in asphyxia is generally corrected soon after birth before significant hyperbilirubinemia develops in preterm infants. Although one study from Pakistan showed birth asphyxia was a risk factor for severe jaundice (Aynalem et al., 2020).

Review intrapartal records for specific risk factors, such as low birth weight (LBW) or intrauterine growth restriction (IUGR), prematurity, abnormal metabolic processes, vascular injuries, abnormal circulation, sepsis , or polycythemia. Certain clinical conditions may cause a reversal of the blood-brain barrier, allowing bound bilirubin to separate either at the cell membrane level or within the cell itself, increasing the risk of CNS involvement. The higher the blood bilirubin level is, the deeper jaundice and the greater the risk for neurological damage.

Observe the infant on the sclera and oral mucosa, yellowing of skin immediately after blanching, and specific body parts. Assess oral mucosa, posterior portion of the hard palate , and conjunctival sacs in dark-skinned newborns. The yellow discoloration of the skin and sclera in neonates diagnosed with jaundice results from the accumulation of unconjugated bilirubin. Neonatal jaundice first becomes visible on the face and forehead (Hansen & Aslam, 2017). Clinical appearance of jaundice is evident at bilirubin levels >7–8 mg/dl in full-term infants. Note: Yellow underlying pigment may be normal in dark-skinned infants.

Evaluate maternal and prenatal nutritional levels; note possible neonatal hypoproteinemia, especially in preterm infants. Hypoproteinemia in the newborn may result in jaundice. One gram of albumin carries 16 mg of unconjugated bilirubin. Lack of sufficient albumin increases the amount of unbound circulating (indirect) bilirubin, which may cross the blood-brain barrier. The binding of compounds to albumin may reduce their toxicity, such as in the case of unconjugated bilirubin in the neonate. Albumin is also involved in maintaining acid-base balance as it acts as a plasma buffer (Gounden et al., 2021).

Note infant’s age at onset of jaundice; differentiate the type of jaundice (i.e., physiological, breast milk–induced, or pathological). Physiological jaundice usually appears between the 2nd and 3rd days of life, as excess RBCs needed to maintain adequate oxygenation for the fetus are no longer required in the newborn and are hemolyzed, thereby releasing bilirubin, the final breakdown product of heme. Breast milk jaundice usually appears between the 4th and 6th days of life, affecting only 1%–2% of breastfed infants. Some women’s breast milk is thought to contain an enzyme (pregnanediol) that inhibits glucuronyl transferase (the liver enzyme that conjugates bilirubin) or contain several times the normal breast milk concentration of certain free freezer fatty acids, which are also thought to inhibit the conjugation of bilirubin. Pathological jaundice appears within the first 24 hr of life and is more likely to lead to the development of kernicterus/bilirubin encephalopathy.

Assess infant for progression of signs and behavioral changes. Excessive unconjugated bilirubin (associated with pathologic jaundice) has an affinity for extravascular tissue, including the basal ganglia of brain tissue. Behavior changes associated with kernicterus usually occur between the 3rd and 10th days of life and rarely occur before 36 hours of life. The characteristic clinical manifestations of kernicterus that are routinely described and are consistent with neuropathological findings include athetoid cerebral palsy , paralysis of upward gaze, and hearing disorders. However, these may represent only “the tip of the iceberg” (Amin et al., 2018).

Evaluate infant for pallor, edema , or hepatosplenomegaly These signs may be associated with hydrops fetalis, Rh incompatibility, and in utero hemolysis of fetal RBCs. With Rh incompatibility, an infant may not appear pale at birth despite the red cell destruction that occurred in utero because the accelerated production of red cells during the last few months in utero compensates to some degree for the destruction. The liver and spleen may be enlarged from attempts to destroy damaged blood cells. Suppose the number of red cells has significantly decreased. In that case, the blood in the vascular circulation may be hypotonic to interstitial fluid, causing fluid to shift from the lower to higher isotonic pressure by osmosis , resulting in extreme edema . Hydrops fetalis is a Greek term that refers to a pathologic accumulation of at least two or more cavities with a fluid collection in the fetus.

Assess the neonate’s bilirubin blood levels regularly. Phototherapy success is determined by frequently measuring serum bilirubin levels. Neonatal hyperbilirubinemia is extremely common because almost every newborn develops an unconjugated serum bilirubin level of more than 1.8 mg/dL during the first week of life. Significant jaundice was defined according to gestational and postnatal age and leveled off at 14 mg/dL at four days in preterm infants and 17 mg/dL in the term infants (Hansen & Aslam, 2017).

Assess infant for signs of hypoglycemia . Hypoglycemia necessitates fat stores for energy-releasing fatty acids, which compete with bilirubin for binding sites on albumin. A study reported that 70.8% of late preterm neonates and 29.1% of term neonates has at least one neonatal morbidity like neonatal jaundice, hypoglycemia , respiratory morbidities, and sepsis . They observed jaundice in 55.1% of late preterm neonates who required phototherapy, and hypoglycemia was found in 8.8% of late preterm neonates (Salman et al., 2021).

Initiate early oral feedings within 4–6 hours following birth, especially if the infant is breastfed. This establishes proper intestinal flora necessary for reducing bilirubin to urobilinogen; decreases the enterohepatic circulation of bilirubin (bypassing the liver with the persistence of ductus venosus), and decreases reabsorption of bilirubin from the bowel by promoting passage of meconium . A delay in enteral feeding may limit intestinal motility and bacterial colonization, resulting in decreased bilirubin clearance (Aynalem et al., 2020).

Keep infant warm and dry; frequently monitor skin and core temperature. Cold stress potentiates the release of fatty acids, which compete for binding sites on albumin, thereby increasing freely circulating (unbound) bilirubin. A neutral thermal environment permits the infant to maintain a normal core temperature with minimum oxygen consumption and caloric expenditure. Preterm infants have little or no muscular activity; they remain in an extended posture because of a lack of muscle tone; they cannot shiver.

Apply transcutaneous jaundice meter. Since visual assessment of jaundice is not accurate, both the American Academy of Pediatrics and the Spanish Association of Pediatrics recommend that all newborns as of 35 weeks of gestation undergo screening for hyperbilirubinemia by measuring either total serum bilirubin (SB) or transcutaneous bilirubin (TcB). Transcutaneous bilirubinometry measures the bilirubin subcutaneously, and therefore, TcB is not the same value as SB. although current jaundice meters have been designed to agree as closely as possible with SB (Maya-Enero et al., 2021).

Discontinue breastfeeding for 24–48 hr, as indicated. Assist mother as needed with the pumping of breasts and reestablishment of breastfeeding. Opinions vary as to whether interrupting breastfeeding is necessary when jaundice occurs. However, formula ingestion increases GI motility and excretion of stool and bile pigment, and serum bilirubin levels begin to fall within 48 hours after discontinuation of breastfeeding. Certain factors present in the breast milk of some mothers may also contribute to the increased enterohepatic circulation of bilirubin (breast milk jaundice). Beta-glucuronidase may play a role by uncoupling bilirubin from its binding to glucuronic acid, thus making it available for reabsorption (Hansen & Aslam, 2017).

Monitor laboratory studies, as indicated . See Diagnostic and Laboratory Procedures

Calculate plasma bilirubin-albumin binding capacity. This aids in determining the risk of kernicterus and treatment needs. When the total bilirubin value divided by total serum protein level is <3.7, the danger of kernicterus is very low. However, the risk of injury is dependent on the degree of prematurity, presence of hypoxia or acidosis, and drug regimen (e.g., sulfonamides , chloramphenicol ) (Hansen & Aslam, 2017).

Initiate phototherapy per protocol, using fluorescent bulbs above the infant or bile blanket (except for newborns with Rh disease). Phototherapy causes photooxidation of bilirubin in subcutaneous tissue, thereby increasing the water solubility of bilirubin, which allows rapid excretion of bilirubin in stool and urine . The rate of bilirubin reduction is related to phototherapy, so an exchange transfusion is the only appropriate treatment. Phototherapy is discontinued when the bilirubin level steadily declines to 14 mg/dL.

Administer enzyme induction agent  ( phenobarbital , ethanol) as appropriate. Medications are not usually administered in infants diagnosed with physiologic neonatal jaundice. However, in certain instances, phenobarbital, an inducer of hepatic bilirubin metabolism, has been used to enhance bilirubin metabolism. Several studies have shown that phenobarbital effectively reduces mean serum bilirubin values during the first week of life (Hansen & Aslam, 2017).

Assist with preparation and administration of exchange transfusion. Exchange transfusion can be used as therapy for blood incompatibility, wherein it removes approximately 85% of sensitized red cells. It reduces the serum concentration of indirect bilirubin and can prevent heart failure in infants with severe anemia or polycythemia. The type of blood used for transfusion is O Rh-negative blood, even if an infant’s blood type is positive; if Rh-positive or type A or B blood was given, the maternal antibodies that entered the infant’s circulation would destroy this blood also, and the transfusion would be ineffective.

Note the condition of the infant’s cord before transfusion if the umbilical vein is to be used. If the cord is dry, administer saline soaks for 30–60 min before the procedure. Soaks may be necessary to soften the cord and umbilical vein before transfusion for IV access and ease the umbilical catheter’s passage. In general, access via the umbilical vein is the recommended exchange transfusion method for treating severe hyperbilirubinemia in neonates. However, some reports have shown that exchange transfusion via the umbilical vessels is a relatively high-risk procedure (Chen et al., 2008).

Verify infant’s and mother’s blood type and Rh factor. Note blood type and Rh factor of blood to be exchanged. Exchange transfusions are most often associated with Rh incompatibility problems. Using Rho(D)-positive blood would only increase hemolysis and bilirubin levels because antibodies in an infant’s circulation would destroy new RBCs. The type of blood used for transfusion is O Rh-negative blood, even if an infant’s blood type is positive.

Assess the infant’s weight prior to transfusion and for consequent weight changes. Adverse events are more frequent in infants of lower gestation and birth weights and those who were sicker. These results are similar to other studies because sicker and smaller infants have multiple comorbidities and are at risk of more complications (Chacham et al., 2019). Additionally, weight change reveals weight gain related to fluid overload . Fluid overload can cause respiratory and cardiac complications.

Assess the infant for neurologic changes. Irritability, twitching, convulsions, or seizures are signs of neurotoxicity resulting from jaundice. An increasing bilirubin level becomes dangerous if the level rises above 20 mg/dL in a term infant and perhaps as low as 12 mg/dL in a preterm infant because brain damage from bilirubin-induced neurologic dysfunction (BIND), a wide spectrum of disorders caused by increasingly severe hyperbilirubinemia ranging from mild dysfunction to acute bilirubin encephalopathy (ABE) (invasion of bilirubin into brain cells), can occur.

Assess infant for excessive bleeding from IV site following the transfusion. Infusion of heparinized blood (or citrated blood without calcium replacement) alters coagulation for 4–6 hr following the exchange transfusion and may result in bleeding . Thrombocytopenia was seen in 57.4% of neonates undergoing exchange transfusion. It was observed that there was a decrease in the platelet count following the transfusion with a nadir at 24 hours and full recovery at or after 72 hours (Chacham et al., 2019).

Monitor venous pressure, pulse, color, and respiratory rate/ease before, during, and after transfusion. Suction as needed. This establishes baseline values, identifies potentially unstable conditions (e.g., apnea or cardiac dysrhythmia /arrest), and maintains the airway. Exchange transfusion is not free of risk, with the estimated morbidity rate at 5% and the mortality rate as high as 0.5%. The most common adverse events are apnea , bradycardia, cyanosis , vasospasm, and hypothermia with metabolic abnormalities (Wagle & Aslam, 2017). Bradycardia may occur if calcium is injected too rapidly. 

Monitor for signs of electrolyte imbalance (e.g., lethargy , seizure activity, and apnea ;  hyperreflexia, bradycardia, or diarrhea ). Hypocalcemia and hyperkalemia may develop during and following exchange transfusion. Hypocalcemia is one of the most frequent adverse events, with the incidence ranging from 22.5% to 98%. Hypocalcemia following exchange transfusion is due to certain chelating properties of citrate, which is present in a very high concentration in the donor blood as a component of the anticoagulant (Chacham et al., 2019).

Assess the infant for any congenital diseases such as other hemolytic diseases and cardiac failure. An infant born with cardiac failure and edema resulting from hemolytic disease is a candidate for immediate exchange transfusion with fresh whole blood. When red cells have significantly decreased due to a hemolytic disease, the blood in the vascular circulation may be hypotonic to interstitial fluid, causing fluid to shift from lower to higher isotonic pressure by osmosis in extreme edema.

Maintain the infant’s temperature prior to, during, and after the procedure. Place infant under radiant warmer with servomechanism. A transfusion should be done under a radiant heat warmer to keep the infant warm during what can be a lengthy procedure to prevent energy expenditure from having to maintain body temperature. This also helps prevent vasospasm, reduces the risk of ventricular fibrillation , and decreases blood viscosity.

Warm the blood prior to infusion by placing it in blood warmer. Donor blood must be maintained at room temperature, or hypothermia from the cold insult could result. Use only commercial blood warmers to warm the blood, not hot towels or a radiant heat warmer, to avoid destroying red cells.

Ensure freshness of blood (not more than two days old), with heparinized blood preferred. Older blood is more likely to hemolyze, thereby increasing bilirubin levels. Moreover, old stored blood has high levels of leucocyte-secreted cytokines that significantly raise the risk of non-hemolytic febrile transfusion reactions. Heparinized blood is always fresh but must be discarded if not used within 24 hr. Blood transfusion using heparinized blood can be employed to avoid citrate toxicity in neonates undergoing exchange transfusion. Moreover, repetitive small top-up transfusions can also be carried out with fresh heparinized blood collected as and when needed from a dedicated safe walking donor (Ahmed & Ibrahim, 2018).

Avoid overheating of blood prior to transfusion. Too much heat in the blood promotes hemolysis and the release of potassium , causing hyperkalemia. The risk of hemolysis with heated blood requires the choice of heating temperature to be considered. In their study, Van der Walt and Russel (Van der Walt & Russel, 1978) argue that the blood should ideally be heated to a human body temperature of 37°C (98.6℉), but that any temperature between 32°C (89.6℉) and 37°C (98.6℉) is acceptable (Poder et al., 2015).

Ensure availability of resuscitative equipment. Access to resuscitative equipment provides immediate support if necessary. Exchange transfusions can lead to complications such as life-threatening bleeding, sepsis, cardiac arrhythmias, and even death, apart from transient hypocalcemia , hyperkalemia, bradycardia, and thrombocytopenia (Chacham et al., 2019).

Maintain NPO status for 4 hr prior to the procedure, or aspirate gastric contents. The infant should be nil orally as soon as the decision is made to perform an exchange transfusion. Pass an oro- nasogastric tube and aspirate stomach contents. Leave the tube in situ and on free drainage for the duration of the procedure (The Royal Children’s Hospital, 2004). This reduces the risk of possible regurgitation and aspiration during the procedure.

Carefully document events during transfusion, recording the amount of blood withdrawn and injected (usually 7–20 ml at a time). Documentation helps prevent errors in fluid replacement. The amount of blood exchanged is approximately 170 ml/kg of body weight. A double-volume exchange transfusion ensures that between 75% and 90% of circulating RBCs are replaced. The process is time-consuming and labor -intensive but remains the ultimate treatment to prevent kernicterus (Wagle & Aslam, 2017).

Administer albumin prior to transfusion if indicated. Although somewhat controversial, administration of albumin may increase the albumin available for bilirubin binding, thereby reducing levels of freely circulating serum bilirubin. Synthetic albumin is not thought to increase available binding sites. Due to the elicited increase in plasma bilirubin, albumin administration to reduce bilirubin-induced neurological damage invalidates the use of plasma total bilirubin as an indicator of the overall risk of bilirubin neurotoxicity (Vodret et al., 2015).

Administer medications, as indicated . See Pharmacologic Management

Administer antibiotics as indicated. Antibiotics prevent and/or treat infections. After a transfusion, the infant is closely observed to be certain that there is no umbilical vessel inflammation of the cord if this was the transfusion site, which would suggest infection .

Assist with administration of intravenous immunoglobulin (IVIG) as indicated. IVIG has been shown to reduce the need for exchange transfusion in hemolytic disease of the newborn due to Rh or ABO incompatibility. The number needed to treat to prevent one exchange transfusion was noted to be 2.7 and was estimated to be ten if all the infants with strongly positive direct Coombs test were to receive the medication . Although IVIG has been proven safe, a retrospective review reported an almost 30-times increased risk of necrotizing enterocolitis in late preterm and term infants (Wagle & Aslam, 2017).

Note the presence or development of biliary or intestinal obstruction. Phototherapy is contraindicated in these conditions because the photoisomers of bilirubin produced in the skin and subcutaneous tissues by exposure to light therapy cannot be readily excreted. The risk of secondary intestinal obstruction may increase after phototherapy. The velocity of blood flow in the upper mesenteric artery at the end of the diastolic period is accelerated post-phototherapy, indicating that the mesenteric vascular smooth muscle may undergo diastolic changes during phototherapy, leading to mesenteric ischemia , which may be one of the causes of intestinal obstruction in premature infants (Wang et al., 2021).

Monitor the neonate’s skin and core temperature every two hours or more frequently until stable. Regulate incubator/ Isolette temperature as appropriate. Fluctuations in body temperature can occur in response to light exposure, radiation , and convection. When the jaundiced newborn is treated with blue phototherapy, apart from the areas protected by the black blindfold and the diaper, all other areas are exposed to illumination. As a result, neonates diagnosed with jaundice treated with blue light often experience alterations in body temperature (Wang et al., 2021).

Note color and frequency of stools and urine. Frequent, greenish, loose stools and greenish urine indicate the effectiveness of phototherapy with the breakdown and excretion of bilirubin. The nurse must determine loose, greenish stools caused by photodegradation products from true diarrhea .

Monitor fluid intake and output; weigh infant twice a day. Note signs of dehydration (e.g., reduced urine output, depressed fontanels , dry or warm skin with poor turgor, and sunken eyes). Dehydration may occur during phototherapy, particularly in premature infants. By measuring the skin moisture content of premature infants before and after phototherapy, Maayan-Metzger et al. found that the mean skin moisture loss increased by 26.4% during phototherapy, with the most significant loss observed in the elbow socket, groin, and back (Wang et al., 2021). Note: Infant may sleep for longer periods in conjunction with phototherapy, increasing the risk of dehydration if a frequent feeding schedule is not maintained.

Evaluate the appearance of skin and urine, noting brownish-black color. An uncommon side effect of phototherapy involves exaggerated pigment changes (bronze baby syndrome), which may occur if conjugated bilirubin levels rise. The changes in skin color may last for 2–4 months but are not associated with harmful sequelae. The bronze baby syndrome is an irregular pigmentation resulting from phototherapy in newborn infants diagnosed with neonatal jaundice that is mainly noticeable in the skin, mucous membranes, and urine and generally occurs in neonates with elevated serum conjugated bilirubin levels (Wang et al., 2021).

Note behavioral changes or signs of deteriorating condition (e.g., lethargy , hypotonia, hypertonicity, or extrapyramidal signs). Such changes may indicate the deposition of bile pigment in the basal ganglia and developing kernicterus. Following neonatal phototherapy, the serum level of total free calcium is often diminished, leading to hypocalcemia, which is higher among premature infants than that among full-term infants (Wang et al., 2021).

Assess for the presence of rash and petechiae. Certain newborns develop petechiae and skin rashes from phototherapy, which gradually fade when phototherapy is discontinued. Petechiae may be associated with light-induced thrombocytopenia; thus, the platelet count should be closely monitored during phototherapy. A small number of infants diagnosed with cholestatic jaundice develop a purpuric rash and bullous eruptions after phototherapy, which may increase the total circulating porphyrin levels (Wang et al., 2021).

Note fussiness or increased crying episodes and irritability. It has been reported that newborns receiving phototherapy have more frequent crying episodes than those receiving no therapy for clinical jaundice, which may be associated with changes in the circadian rhythm during neonatal phototherapy.

Document the type of fluorescent lamp, the total number of hours since bulb replacement, and the measured distance between lamp surface and infant. Light emission may decay over time. The infant should be approximately 18–20 in (45 cm) from the light source for maximal benefit. Note: A fiberoptic blanket connected to an illuminator (light source) allows the infant to be “wrapped” in therapeutic light without risk to corneas. In addition, infants can be held and fed without interrupting therapy.

Measure the quantity of photon energy of fluorescent bulbs (white or blue light) using a photometer. The intensity of light striking the skin surface from the blue spectrum (blue lights) determines how close to the light source the infant should be placed. The photometer should register between 8 and 10 mW/cm 2 /nm of light when placed flush with the infant’s abdomen. Blue and special blue lights are considered more effective than white light in promoting bilirubin breakdown, but they create difficulty in evaluating the newborn for cyanosis. The American Academy of Pediatrics defines standard phototherapy as 8-10 mW/cm 2 per nm and intensive phototherapy as more than 30 mW/cm 2 per nm in the 430-490 nm band (Sawyer & Nimavat, 2018).

Cover the testes and penis of a male infant. The infant is undressed except for a diaper to protect the ovaries or testes, and so as much skin surface as possible is exposed to light. Some phototherapy lights may affect reproduction. Potential and embryonic development because of the combined effects of light penetration in the tissues. The light had probably stimulated some neuroendocrine structures in the skin. Koc et al. reported the seminiferous tubule diameters were thinner than the control group after phototherapy in rats. Similar histological changes were obtained in cryptorchid testis in humans (Cetinkursun et al., 2006).

Apply patches to closed eyes; inspect eyes every two hours when patches are removed for feedings. Monitor placement frequently. Retinal damage represents another challenge associated with phototherapy for neonatal jaundice. The light-sensitive retinas absorb photons more readily when exposed to blue light, most effective at degrading bilirubin. Following continuous or stronger blue light irradiation, the retinal function degenerates due to a significantly increased retinal cell death rate (Wang et al., 2021). In addition to eye shields, many centers also prescribe lubricating eye drops for an infant receiving phototherapy (Sawyer & Nimavat, 2018).

Cleanse the infant’s eyes using sterile or normal saline water. As the incidence of conjunctivitis is increased among children receiving phototherapy who wear eye masks over prolonged periods, thorough eye care, such as cleaning eye secretions and surrounding skin with normal saline cotton balls, must be applied (Wang et al., 2021). 

Reposition the infant every two hours. This allows equal exposure of skin surfaces to fluorescent light, prevents excessive exposure of individual body parts, and limits pressure areas.

Carefully wash the perianal area after each passage of stool; inspect the skin for possible irritation or breakdown. Early intervention helps prevent irritation and excoriation from frequent or loose stools. An infant’s stools under bilirubin lights are often bright green because of the excessive bilirubin being excreted as a result of the therapy. They are also frequently loose and may be irritating to the skin.

Encourage an increased oral fluid intake. To prevent the loss of water and electrolytes caused by phototherapy, water and electrolytes must be replenished when necessary. The warming effect of conventional phototherapy increases water loss from the body surface, while light-emitting diode (LED) phototherapy, which is currently widely used, causes less water loss (Wang et al., 2021).

Bring infant to parents for feedings. Encourage stroking, cuddling, eye contact, and talking to the infant during feedings. Encourage parents to interact with the infant in the nursery between feedings. This fosters the attachment process, which may be delayed if phototherapy requires separation. Visual, tactile, and auditory stimulation helps the infant overcome sensory deprivation. Intermittent phototherapy does not negatively affect the photooxidation process. Note: Dependent on infant condition and policies/capabilities of the hospital, phototherapy may be provided in conjunction with rooming-in.

Ensure that the infant’s chest is properly shielded during phototherapy. 50% of premature infants receiving phototherapy were diagnosed with patent ductus arteriosus, the re-opening of the ductus arteriosus may be evoked by blue light penetrating the chest wall of the premature infant and causing relaxation of the smooth muscle of the cardiovascular system by activating the Ca2+ dependent K+ channel. It has been reported that appropriate shielding of the chest during phototherapy may reduce the incidence of patent ductus arteriosus (Wang et al., 2021).

Administering medications and providing pharmacologic support in patients with hyperbilirubinemia (jaundice) involves careful consideration of the specific medications used, their dosages, and their potential effects on bilirubin metabolism and liver function.

  • 10% calcium gluconate . From 2–4 ml of calcium, gluconate may be administered after every 100 ml of blood infusion to correct hypocalcemia and minimize possible cardiac irritability (Wani et al., 2018). The general approach is to give IV calcium during exchange transfusion, but this issue is still controversial. There is not enough literature data about calcium requirements during exchange transfusion. Complications such as tetany convulsions and death have been reported when blood products containing adenine-citrate dextrose (ACD) were used for exchange transfusion despite IV calcium infusion (Aydin et al., 2021).
  • Sodium bicarbonate . Sodium bicarbonate corrects acidosis. Relatively higher serum pH may be attributed to higher serum bicarbonate concentration in freshly CPDA-treated blood. The serum bicarbonate concentration in CPDA-treated blood usually starts decreasing after three days of storage.
  • Protamine sulfate . Protamin sulfate counteracts anticoagulant effects of heparinized blood. Some clinicians remain skeptical about the safety of heparinized blood transfusion due to the possible risk of heparin -induced bleeding. This skepticism may be valid for hemostatically unstable neonates due to comorbid hemorrhagic conditions such as hemophilia , thrombocytopenia, DIC, or vitamin K deficiency. If overheparinization is clinically suspected during or after the transfusion, protamine sulfate can be used to neutralize it (Ahmed & Ibrahim, 2018).
  • Administer enteral or parenteral fluid as indicated. Fluids compensate for insensible and intestinal fluid losses and supply nutrients if feedings are withheld during phototherapy for infants with severe hyperbilirubinemia. Due to this increase in insensible water loss, recommendations have been made to increase maintenance fluid by 10 ml/kg/day in premature infants exposed to conventional phototherapy (Sawyer & Nimavat, 2018).

Monitoring the results of diagnostic and laboratory procedures is crucial in the management of hyperbilirubinemia (jaundice) to assess the severity of the condition, identify the underlying cause, and guide appropriate treatment decisions. Various diagnostic tests and laboratory procedures are employed to evaluate liver function, bilirubin levels, and other relevant parameters.

  • Direct and indirect bilirubin . Bilirubin appears in two forms: direct bilirubin, which is conjugated by the liver enzyme glucuronyl transferase, and indirect bilirubin, which is unconjugated and appears in a free form in the blood or bound to albumin. Elevated levels of indirect bilirubin best predict the infant’s potential for kernicterus. Elevated indirect bilirubin levels of 18–20 mg/dl in the full-term infant or13–15 mg/dl in preterm or sick infants are significant (Hansen & Aslam, 2017).
  • Total serum bilirubin level . Usually, a total serum bilirubin level test is the only one required in an infant with moderate jaundice who presents on the typical second or third day of life without a history or physical findings suggestive of a pathologic process (Hansen & Aslam, 2017).
  • Direct/indirect Coombs’ test on cord blood . Positive results of the indirect Coombs test indicate the presence of antibodies (Rh-positive or anti-A, anti-B) in the mother’s and newborn’s blood; positive results of the direct Coombs test indicate the presence of sensitized (Rh-positive, anti-A, or anti-B) RBCs in the neonate.
  • CO2-combining power; Reticulocyte count and peripheral smear . A decrease is consistent with hemolysis. Excessive hemolysis causes reticulocyte count to increase. Smear identifies abnormal or immature RBCs. The reticulocyte count provides an indirect insight into the bone marrow condition by distinguishing if the anemia is related to inadequate RBC production or accelerated loss/destruction (Szigeti & Staros, 2014).
  • Hemoglobin /hematocrit (Hb/Hct) . Elevated Hb/Hct levels (Hb 22 g/dl; Hct 65%) indicate polycythemia, possibly caused by delayed cord clamping, maternal-fetal transfusion, twin-to-twin transfusion, maternal diabetes , or chronic intrauterine stress and hypoxia, as seen in low birth weight (LBW) infant or infant with compromised placental circulation. Hemolysis of excess RBCs causes elevated bilirubin levels with 1 g of Hb yielding 35 mg of bilirubin. Low Hb levels (14 mg/dl) may be associated with hydrops fetalis or Rh incompatibility occurring in utero and causing hemolysis, edema, and pallor.
  • Total serum protein or serum albumin levels . Low serum protein levels (3.0 g/dl) indicate a reduced binding capacity for bilirubin. Serum albumin levels appear to be a useful adjunct in evaluating the risk of toxicity levels because albumin binds bilirubin in a ratio of 1:1 at the primary high-affinity binding site (Hansen & Aslam, 2017).
  • Serum calcium and potassium . Measure urea and electrolytes regularly until the infant is stable, as indicated. Donor blood containing citrate as an anticoagulant binds calcium, decreasing serum calcium levels. In addition, if blood is more than two days old, RBC destruction releases potassium, creating a risk of hyperkalemia and cardiac arrest (The Royal Children’s Hospital, 2004).
  • Glucose . Perform blood glucose levels immediately post-procedure and then hourly until the infant is stable (The Royal Children’s Hospital, 2004). There is a significant rise in post-exchange serum glucose , attributed to high dextrose content in the donor blood and anticoagulants for preservation. Although normal mean blood sugar was observed at 12 hours post-exchange, one needs to be cautious as rebound hypoglycemia may occur during the initial hours (Wani et al., 2018).
  • Serum pH levels. The serum pH of donor blood is typically 6.8 or less. Acidosis may result when fresh blood is not used, and the infant’s liver cannot metabolize citrate used as an anticoagulant, or when donor blood continues anaerobic glycolysis with the production of acid metabolites. Low serum potassium after exchange transfusion coincided with higher serum pH, which is known to cause a shift of serum potassium into the intracellular compartment (Wani et al., 2018).
  • Platelets and WBCs . Thrombocytopenia during phototherapy has been reported in some infants. A decrease in WBCs suggests a possible effect on peripheral lymphocytes . There is a significant association between the decrease in platelet count with the duration of phototherapy and lower gestational age in the neonate (Sarkar et al., 2021).
  • Riboflavin levels . Since the wavelength of absorption of blue light by riboflavin is similar to that of bilirubin, both riboflavin and bilirubin will decompose at the same time when a newborn with jaundice receives blue light therapy, leading to the loss of riboflavin in the body. The riboflavin deficiency will reduce the synthesis of active riboflavin adenine dinucleotide, impair hydrogen delivery of erythrocytes , reduce glutathione reductase, and weaken the activity of erythrocyte glutathione reductase, thus aggravating hemolysis (Wang et al., 2021).

Recommended nursing diagnosis and nursing care plan books and resources.

Disclosure: Included below are affiliate links from Amazon at no additional cost from you. We may earn a small commission from your purchase. For more information, check out our privacy policy .

Ackley and Ladwig’s Nursing Diagnosis Handbook: An Evidence-Based Guide to Planning Care We love this book because of its evidence-based approach to nursing interventions. This care plan handbook uses an easy, three-step system to guide you through client assessment, nursing diagnosis, and care planning. Includes step-by-step instructions showing how to implement care and evaluate outcomes, and help you build skills in diagnostic reasoning and critical thinking.

case study neonatal hyperbilirubinemia

Nursing Care Plans – Nursing Diagnosis & Intervention (10th Edition) Includes over two hundred care plans that reflect the most recent evidence-based guidelines. New to this edition are ICNP diagnoses, care plans on LGBTQ health issues, and on electrolytes and acid-base balance.

case study neonatal hyperbilirubinemia

Nurse’s Pocket Guide: Diagnoses, Prioritized Interventions, and Rationales Quick-reference tool includes all you need to identify the correct diagnoses for efficient patient care planning. The sixteenth edition includes the most recent nursing diagnoses and interventions and an alphabetized listing of nursing diagnoses covering more than 400 disorders.

case study neonatal hyperbilirubinemia

Nursing Diagnosis Manual: Planning, Individualizing, and Documenting Client Care  Identify interventions to plan, individualize, and document care for more than 800 diseases and disorders. Only in the Nursing Diagnosis Manual will you find for each diagnosis subjectively and objectively – sample clinical applications, prioritized action/interventions with rationales – a documentation section, and much more!

case study neonatal hyperbilirubinemia

All-in-One Nursing Care Planning Resource – E-Book: Medical-Surgical, Pediatric, Maternity, and Psychiatric-Mental Health   Includes over 100 care plans for medical-surgical, maternity/OB, pediatrics, and psychiatric and mental health. Interprofessional “patient problems” focus familiarizes you with how to speak to patients.

case study neonatal hyperbilirubinemia

Other recommended site resources for this nursing care plan:

  • Nursing Care Plans (NCP): Ultimate Guide and Database MUST READ! Over 150+ nursing care plans for different diseases and conditions. Includes our easy-to-follow guide on how to create nursing care plans from scratch.
  • Nursing Diagnosis Guide and List: All You Need to Know to Master Diagnosing Our comprehensive guide on how to create and write diagnostic labels. Includes detailed nursing care plan guides for common nursing diagnostic labels.

Other care plans related to the care of the pregnant mother and her baby:

  • Abortion (Termination of Pregnancy) | 8 Care Plans
  • Cervical Insufficiency (Premature Dilation of the Cervix) | 4 Care Plans
  • Cesarean Birth | 11 Care Plans
  • Cleft Palate and Cleft Lip | 7 Care Plans
  • Gestational Diabetes Mellitus | 8 Care Plans
  • Hyperbilirubinemia (Jaundice) | 4 Care Plans
  • Labor Stages, Induced, Augmented, Dysfunctional, Precipitous Labor | 45 Care Plans
  • Neonatal Sepsis | 8 Care Plans
  • Perinatal Loss (Miscarriage, Stillbirth) | 6 Care Plans
  • Placental Abruption | 4 Care Plans
  • Placenta Previa | 4 Care Plans
  • Postpartum Hemorrhage | 8 Care Plans
  • Postpartum Thrombophlebitis | 5 Care Plans
  • Prenatal Hemorrhage (Bleeding in Pregnancy) | 9 Care Plans
  • Preeclampsia and Gestational Hypertension  | 6 Care Plans
  • Prenatal Infection | 5 Care Plans
  • Preterm Labor | 7 Care Plans
  • Puerperal & Postpartum Infections  | 5 Care Plans
  • Substance Abuse in Pregnancy | 9 Care Plans

Resources and journals you can use to further your reading about Hyperbilirubinemia (Jaundice).

  • Ahmed, S.G., & Ibrahim, U.A. (2018, April). Donor Blood Selection Criteria For Neonatal Red Cell Transfusion: General And Tropical Perspectives. The Tropical Journal of Health Sciences , 25 (2).
  • Amin, S. B., Smith, T., & Timler, G. (2018, October 23). Developmental influence of unconjugated hyperbilirubinemia and neurobehavioral disorders . Pediatric Research , 85 , 191-197.
  • Aydin, B., Yilmaz, H. C., Botan, E., Aktepe, A. O., & Dilli, D. (2021, December). Is it necessary to give calcium infusion during the exchange transfusion in newborns? Transfusion and Apheresis Science , 30 (6), 103236.
  • Aynalem, S., Abayneh, M., Metaferia, G., Demissie, A. G., Gidi, N. W., Demtse, A. G., Berta, H., Worku, B., Nigussie, A. K., Mekasha, A., Bonger, Z. T., McClure, E. M., Goldenberg, R. L., & Muhe, L. M. (2020). Hyperbilirubinemia in Preterm Infants Admitted to Neonatal Intensive Care Units in Ethiopia . Global Pediatric Health , 7 , 1-8.
  • Cetinkursun, S., Demirbag, S., Cincik, M., Baykal, B., & Gunal, A. (2006, January-February). Effects of Phototherapy on Newborn Rat Testicles . Archives of Andrology , 52 (1), 61-70.
  • Chacham, S., Kumar, J., Dutta, S., & Kumar, P. (2019, April-June). Adverse Events Following Blood Exchange Transfusion for Neonatal Hyperbilirubinemia: A Prospective Study. Journal of Clinical Neonatology , 8 (2), 79-84.
  • Chen, H.-N., Lee, M.-L., & Tsao, L.-Y. (2008, October). Exchange Transfusion Using Peripheral Vessels Is Safe and Effective in Newborn Infants. Pediatrics , 122 (4).
  • Gounden, V., Vashisht, R., & Jialal, I. (2021, September 28). Hypoalbuminemia – StatPearls . NCBI. Retrieved May 15, 2022.
  • Hansen, T. W., & Aslam, M. (2017, December 27). Neonatal Jaundice: Background, Pathophysiology, Etiology . Medscape Reference. Retrieved May 15, 2022.
  • Kaplan, M., Zimmerman, D., Shoob, H., & Stein-Zamir, C. (2019, November 19). Post-discharge neonatal hyperbilirubinemia surveillance . Acta Pediatrica , 109 (5), 923-929.
  • Kim, M.-S., Chung, Y., Kim, H., Ko, D.-H., Jung, E., Lee, B. S., Hwang, S.-H., Oh, H.-B., Kim, E. A.-R., & Kim, K.-S. (2020). Neonatal exchange transfusion: Experience in Korea . Transfusion and Apheresis Science , 59 .
  • Koc, H., Altunhan, H., Dilsiz, A., Kaymakci, A., Duman, S., Oran, B., & Erkul, I. (1999, July). Testicular Changes in Newborn Rats Exposed to Phototherapy . Pediatric and Developmental Pathology , 2 (4), 333-336.
  • Leifer, G. (2018). Introduction to Maternity and Pediatric Nursing . Elsevier.
  • Maayan-Metzger, A., Yosipovitch, G., Hadad, E., & Sirota, L. (2001). Transepidermal Water Loss and Skin Hydration in Preterm Infants During Phototherapy . American Journal of Perinatology , 18 (7), 393-396.
  • Maya-Enero, S., Candel-Pau, J., Garcia-Garcia, J., Duran-Jorda, X., & Lopez-Vilchez, M. A. (2021, January 6). Reliability of transcutaneous bilirubin determination based on skin color determined by a neonatal skin color scale of our own . European Journal of Pediatrics , 180 , 607-616.
  • Morrison, K. L. (2021). Improving the Identification of Newborns at Risk for Hyperbilirubinemia. ProQuest .
  • Poder, T. G., Nonkani, W. G., & Leponkouo, T. (2015, July). Blood Warming and Hemolysis: A Systematic Review With Meta-Analysis . Transfusion Medicine Reviews , 29 (3), 172-180.
  • The Royal Children’s Hospital. (2004). Exchange Transfusion: Neonatal . The Royal Children’s Hospital. Retrieved May 19, 2022.
  • Salman, M., Rathore, H., Arif, S., Ali, R., Khan, A. A., & Nasir, M. (2021, January 5). Frequency of Immediate Neonatal Complications (Hypoglycemia and Neonatal Jaundice) in Late Preterm and Term Neonates . Cureus. Retrieved May 15, 2022.
  • Sardari, S., Mohammadizadeh, M., & Namnabati, M. (2019, January 19). Efficacy of Home Phototherapy in Neonatal Jaundice . Journal of Comprehensive Pediatrics , 10 (1).
  • Sarkar, S. K., Biswas, B., Laha, S., Sarkar, N., Mondal, M., Angel, J., Dr, V., Abhisek, K., Kumar, V., Acharya, A., Biswas, P., Mal, S., Ghosh, D., & Mukherjee, T. (2021). A study on the effect of phototherapy on platelet count in neonates with unconjugated hyperbilirubinemia: a hospital-based prospective observational study . Asian Journal of Medical Sciences , 12 (5).
  • Sawyer, T. L., & Nimavat, D. J. (2018, May 1). Phototherapy for Jaundice: Background, Indications, Contraindications . Medscape Reference. Retrieved May 20, 2022.
  • Silbert-Flagg, J., & Pillitteri, A. (2018). Maternal & Child Health Nursing: Care of the Childbearing & Childrearing Family . Wolters Kluwer.
  • Szigeti, R. G., & Staros, E. B. (2014, September 5). Reticulocyte Count and Reticulocyte Hemoglobin Content: Reference Range, Interpretation, Collection, and Panels . Medscape Reference. Retrieved May 15, 2022.
  • Van der Walt, J.H., & Russel, W.J. (1978, August). Effect of Heating on the Osmotic Fragility of Stored Blood . British Journal of Anaesthesia , 50 (8), 815-820.
  • Vodret, S., Bortolussi, G., Schreuder, A. B., Jasprova, J., Vitek, L., Verkade, H. J., & Muro, A. F. (2015). Albumin administration prevents neurological damage and death in a mouse model of severe neonatal hyperbilirubinemia . “ – Wiktionary. Retrieved May 19, 2022.
  • Wagle, S., & Aslam, M. (2017, December 28). Hemolytic Disease of the Newborn Treatment & Management: Approach Considerations, Medical Care, Complications . Medscape Reference. Retrieved May 19, 2022.
  • Wang, J., Guo, G., Cai, W.-Q., & Wang, X. (2021, March). Challenges of phototherapy for neonatal hyperbilirubinemia . Experimental and Therapeutic Medicine , 21 (3).
  • Wani, M. I., Nazir, M., Lone, R., Rafiq, M., Ali, S. W., & Charoo, B. A. (2018, October 5). Impact of Double Volume Exchange Transfusion on Biochemical Parameters in Neonatal Hyperbilirubinemia . International Journal of Pediatric Research , 4 (2).

Reviewed and updated by M. Belleza, R.N.

Leave a Comment Cancel reply

Log in using your username and password

  • Search More Search for this keyword Advanced search
  • Latest content
  • For authors
  • Browse by collection
  • BMJ Journals

You are here

  • Volume 14, Issue 6
  • Preventive effect of prenatal maternal oral probiotic supplementation on neonatal jaundice (POPS Study): A protocol for the randomised double-blind placebo-controlled clinical trial
  • Article Text
  • Article info
  • Citation Tools
  • Rapid Responses
  • Article metrics

Download PDF

  • http://orcid.org/0000-0002-9719-6331 Bekalu Kassie Alemu 1 , 2 ,
  • May Wing Lee 1 ,
  • Maran Bo Wah Leung 1 ,
  • Wing Fong Lee 1 ,
  • http://orcid.org/0000-0003-0010-2819 Yao Wang 1 , 3 ,
  • Chi Chiu wang 1 , 4 ,
  • So Ling Lau 1
  • 1 Department of Obstetrics and Gynaecology , Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong , Hong Kong SAR
  • 2 Department of Midwifery , College of Medicine and Health Science, Debre Markos University , Debre Markos , Ethiopia
  • 3 Institute of Health Sciences , The Chinese University , Hong Kong , Hong Kong SAR
  • 4 School of Biomedical Sciences, Joint Laboratory for Reproductive Medicine , The Chinese University , Hong Kong , Hong Kong SAR
  • Correspondence to Dr So Ling Lau; solinglau{at}cuhk.edu.hk

Introduction Neonatal jaundice is a common and life-threatening health problem in neonates due to overaccumulation of circulating unconjugated bilirubin. Gut flora has a potential influence on bilirubin metabolism. The infant gut microbiome is commonly copied from the maternal gut. During pregnancy, due to changes in dietary habits, hormones and body weight, maternal gut dysbiosis is common, which can be stabilised by probiotics supplementation. However, whether probiotic supplements can reach the baby through the mother and reduce the incidence of neonatal jaundice has not been studied yet. Therefore, we aim to evaluate the effect of prenatal maternal probiotic supplementation on the incidence of neonatal jaundice.

Methods and analysis This is a randomised double-blind placebo-controlled clinical trial among 94 pregnant women (47 in each group) in a tertiary hospital in Hong Kong. Voluntary eligible participants will be recruited between 28 and 35 weeks of gestation. Computer-generated randomisation and allocation to either the intervention or control group will be carried out. Participants will take either one sachet of Vivomixx (450 billion colony-forming units per sachet) or a placebo per day until 1 week post partum. Neither the study participants nor researchers will know the randomisation and allocation. The intervention will be initiated at 36 weeks of gestation. Neonatal bilirubin level will be measured to determine the primary outcome (hyperbilirubinaemia) while the metagenomic microbiome profile of breast milk and maternal and infant stool samples as well as pregnancy outcomes will be secondary outcomes. Binary logistic and linear regressions will be carried out to assess the association of the microbiome data with different clinical outcomes.

Ethics and dissemination Ethics approval is obtained from the Joint CUHK-NTEC Clinical Research Ethics Committee, Hong Kong (CREC Ref: 2023.100-T). Findings will be published in peer-reviewed journals and presented at international conferences.

Trial registration number NCT06087874 .

  • Clinical Trial
  • NEONATOLOGY
  • Fetal medicine
  • Maternal medicine
  • Prenatal diagnosis

This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See:  http://creativecommons.org/licenses/by-nc/4.0/ .

https://doi.org/10.1136/bmjopen-2023-083641

Statistics from Altmetric.com

Request permissions.

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

STRENGTHS AND LIMITATIONS OF THIS STUDY

This randomised, double-blind, placebo-controlled superiority trial for reaching the baby through the mother to tackle neonatal jaundice is the first of its kind.

Using multistrain probiotics (Vivomixx with eight strains of beneficial bacteria) known for its safety during pregnancy is one of the strengths of this trial.

Biological samples from the mother and baby (stool and breast milk) will be collected to triangulate the effect of the intervention on clinical outcomes with microbiome data.

The participant with her baby will be appointed for the seventh-day follow-up to collect samples and measure the baby’s transcutaneous bilirubin, and this may increase the dropout.

Introduction

Neonatal jaundice is a yellowish discolouration of the skin and sclera of neonates in the first month of life due to elevated levels of bilirubin in the blood (hyperbilirubinaemia). 1–3 Hyperbilirubinaemia is a total serum bilirubin (TSB) level measurement of more than 5 mg/dL or >85.5 µmol/L in 24 hours and 12 mg/dL or 205 µmol/L after a day. 4 Neonatal jaundice can be physiologic or pathologic in its type. The former usually appears after 24 hours of birth and goes off within 2 weeks of life while the latter arises within a day with a TSB level of more than 15 mg/dL and lasts beyond 14 days. 2 5 Physiological jaundice is associated with different factors such as ethnicity, geographical location, birth season, maternal smoking and pharmacotherapy. 6 It is also associated with low birth weight, 7–9 prematurity and sepsis. 8 Pathological jaundice is mostly caused by glucose-6-phosphate dehydrogenase deficiency, 8 10 ABO incompatibility and rhesus haemolysis. 11 Neonatal jaundice needs comprehensive management to avoid severe complications, such as brain damage. However, due to different barriers, evidence-based management of neonatal jaundice is still challenging. 12 Even though there are certain managements such as phototherapy, they have various impacts on the vision, hearing and alertness of the newborn. It also affects the circadian rhythm of babies and causes dehydration, hypocalcaemia and renal damage. 13

Neonatal jaundice is occurring in varied incidence (about 60%–90%) worldwide. 6 The incidence in North America is about 34%. 14 It is more prevalent among infants of Southeast and Far East Asian mothers and may be up to threefold higher than among whites. 6 15 Among the three main regions in China, the Central South Region has extreme hyperbilirubinaemia. A study in Hong Kong showed about 50%–70% of normal-term infants have the problem. 16 Though the bulk of neonatal jaundice causes is unknown, some of the associated factors include breast milk jaundice, breastfeeding jaundice and glucose-6-phosphate dehydrogenase deficiency as well as surgical disease. 17 It is also hypothesised due to elevated enterohepatic recirculation of unconjugated bilirubin, which is mediated by β-glucuronidase enzyme and bad bacteria in the gut. 18 19 Neonates’ gut flora, which is mainly copied from the mother in the uterus, during childbirth, and through breast feeding has a critical role in bilirubin metabolism by antagonising the activity of the β-glucuronidase enzyme and converting conjugated bilirubin into stercobilinogen (to be excreted via faeces) and urobilinogen (via urine). 20 21 In addition to converting conjugated bilirubin into excretable form, good bacteria in the gut keep the neonate’s gut healthy and increase defecation frequency. Even though the gut microbiome cannot have an impact on the pathological causes of hyperbilirubinaemia such as haemolysis, it may facilitate the excretion of conjugated bacteria. 22 23 Thus, it reduces enterohepatic recirculation. On the contrary, the recirculation of unconjugated bilirubin is facilitated during gut microbiome imbalance (dysbiosis). 24

Preventing gut dysbiosis is very crucial and could be one of the management modalities of neonatal jaundice. 25 The mechanism to modulate the infant’s gut is important. Modulating maternal gut microbiome could end up with beneficial bacteria enriched in breast milk. Our systematic review and meta-analysis proved this innovative approach. 26 The way for this modulation of breast milk is the enteromammary pathway/gut-breast axis, where intestinal dendritic cells and CD18 + cells play an important role in helping beneficial bacteria translocate from the maternal gut to the mammary system. 27–29 The infant gut microbiota is the collection of germs colonising the newborn’s intestine. It is thought to be established by breastfeeding and exposure during birth through the birth canal. 30 31 Bioactive components of human breast milk, oligosaccharides, which are non-digestible carbohydrates forming the third largest solid component in human milk particularly are significantly important to shaping infant gut microbiota by nurturing the beneficial bacteria. 32 Antimicrobial agents in breast milk also inactivate pathogens individually, additively and synergistically. 33 34 In the case of gut dysbiosis, pathogenic micro-organisms will be more abundant and play a role in enhancing the beta-glucuronidase enzyme, which indirectly promotes deconjugation and bilirubin reuptake into the enterohepatic circulation. 21

Supplementing pregnant women with products containing beneficial bacteria (probiotics) can maintain gut health. 21 35 36 Probiotics are supplements composed of live beneficial microorganisms to improve microbial balance, lower intestinal pH, decrease colonisation and invasion by pathogenic organisms, and modify the host immune response, mainly in the gastrointestinal tract. They consist of Saccharomyces boulardii, Streptococcus thermophilus, Lactobacillus and Bifidobacterium species. 37 Administration of probiotics directly to infants is evidenced to prevent and treat neonatal problems including jaundice through maintaining gut health, increasing stool frequency, decreasing beta-glucuronidase enzyme activity, compete with pathogens for food and receptors in the gut. 36 38 39 From lines of literature, we found that probiotic species can modulate the physiology of bilirubin metabolism. 21 Such bacteria include but are not limited to Bifidobacterium , Lactobacillus, Streptococcus and Saccharomyces species.

Bifidobacterium

As compared with non-jaundiced babies, those affected by jaundice had significantly lower levels of Bifidobacterium 39 ( B. adolescentis, B. bifidum and B. longum), 40 41 which imply Bifidobacterium has a bilirubin reduction effect. 40 42 An animal study suggested that faecal beta-glucuronidase enzyme was significantly reduced in animals fed diets containing Bifidobacterium ( B. longum ) supplements. 43 Another study conducted among 53 healthy volunteers confirmed that Bifidobacterium ( B. breve, B. lactis and B. bifidum ) has a suppressive effect on beta-glucuronidase activity. 44 45 B. infantis also has an impact on maintaining mucosal and immune systems, reducing sepsis and increasing defecation frequency. 46 47

Lactobacillus

Probiotic Lactobacillus rhamnosus GG has a significant effect on inhibition of serum bilirubin level and increasing defecation frequency to enhance bilirubin excretion. 48 L. rhamnosus and L. acidophilus are species that could reduce hospitalisation of neonates with hyperbilirubinaemia. 45 Lactobacillus bulgaricus as one of the bifid triple viable has a therapeutic effect on neonatal jaundice. 39 Lactobacillus plantarum has also a significant effect on the reduction of bilirubin and liver enzymes. 41

Saccharomyces boulardii

S. boulardii , in an animal model study, affects preventing bacterial translocation and improvement of intestinal barrier function in obstructive jaundice. 49

Streptococcus

S. thermophilus is also one of the bifid triple viable probiotic components and it is effective in treating neonatal jaundice. 39

Since their action on mucosal adhesion, immune system development and inhibition of pathogens is much better in combination than as a single species, multistrain probiotic products are chosen for better effect. 50 51 Therefore, for this project, Vivomixx, a food supplement that is produced according to food Good Manufacturing Practices and a widely studied product on pregnant women with various outcomes of interest could be used. We have selected Vivomixx from available products in the market like ‘PGut pregnancy probiotics’ 52 and ‘Prenatal-Probiotics’ 53 because of two main reasons. First, Vivomixx is a well-applied probiotic on pregnant women with a null adverse event. 54–61 Second, it contains a majority of probiotic species identified to affect neonatal jaundice/ bilirubin metabolism. 56

This is the first study that aims to investigate the preventive effect of maternal probiotic supplementation on the incidence of neonatal jaundice and bilirubin level, pregnancy outcomes, breast milk, as well as maternal and infant stool microbiome profile.

Methods and analysis

Study design, setting and period.

This is a randomised double-blind placebo-controlled parallel group superiority clinical trial in the Prince of Wales Hospital (PWH), Hong Kong from January 2024 to June 2025. This trial will be with pregnant women randomly assigned to either the probiotic group (Vivomixx with maltose) or the placebo group (maltose only). The trial protocol is reported as per the Standard Protocol Items: Recommendations for Interventional Trials guidelines. 62

Participants

Pregnant women who will attend their antenatal care and plan to deliver in PWH will be invited to participate in this study.

Inclusion criteria

Pregnant women from 18 to 45 years old.

Gestational age between 28 and 35 weeks.

Normal singleton pregnancy.

Not on standing dose of antibiotic treatment during enrolment and initiation of the intervention.

Plan to exclusively breastfeed.

Exclusion criteria

Pregnant women with any fetal abnormality.

Couple with glucose 6-phosphate dehydrogenase enzyme deficiency.

Couple with known rhesus or haemolytic disease history.

Any known risk of developing pathological jaundice.

Plan to give birth at other hospitals than PWH.

Women with any contraindications for breast feeding.

Non-exclusively breastfeeding babies.

Recruitment and randomisation

Pregnant women will be recruited between 28 and 35 weeks in the outpatient clinics in a tertiary-level hospital ( figure 1 ). After confirming the eligibility of pregnant women, the investigator will explain the study by using a printed information sheet that contains the title of the study, purpose, expected length of time for participation, description of all the procedures, confidentiality, voluntary participation, the right to refuse the intervention, withdraw from participation in the clinical trial at any time, contact information and all information will be kept securely. Sufficient time is allowed for their consideration, and all their questions will be answered. Pregnant women will voluntarily sign the written informed consent form ( online supplemental file 1 ). Computer-generated randomisation and allocation will be made by a third party other than the investigators and clinicians to allocate the pregnant women to either probiotic or placebo groups in (1:1) ratio. Neither the study participants nor researchers will know the randomisation and allocation. The intervention will be initiated at 36 weeks of gestation for both study groups. To date, the recruitment has not started yet.

Supplemental material

  • Download figure
  • Open in new tab
  • Download powerpoint

Flow diagram of participant recruitment and follow-up in the POPS trial using Consolidated Standards of Reporting Trials. DOL, days of life; N, total screened for eligibility; n, number of participants in each stage of enrolment, intervention and follow-up, POPS, Perinatal Oral Probiotics Supplementation. 76

Intervention

The probiotic group pregnant women will take one sachet of probiotic (a maltose containing Vivomixx) product orally daily from 36 weeks of gestation up to the seventh day of postpartum while pregnant women in the control group will take one sachet placebo (only maltose) orally daily for the same duration. The best time to take the product is in the morning before breakfast. Each participant in both groups will be advised to dilute a sachet of product provided with a cap of cold water. Detailed information will be provided to each participant with an information sheet prepared in local language. Both active Vivomixx and placebo are similar in colour and taste with the same white sachet package. The probiotic product in this trial is a widely studied supplement among pregnant women for other research outcomes including vaginal microbiota and cytokine secretion, bacterial vaginosis, breast milk beneficial microbiota and cytokines, glycaemic control, neonatal gastrointestinal functional symptoms and gut microbiota as well as immune response. 54–59 Maximum gut microbiome colonisation is achieved in 2–3 weeks of supplementation. 63

Investigators have trial identified about 12 different microbiome species from the literature that have direct or indirect bilirubin reduction effects ( table 1 ). There are commercially available probiotic products that contain many of these species. Vivomixx is one of the accessible well-studied products for its safety during pregnancy. It is a non-GMO, gluten-free, high-potency microbiotic food supplement, containing eight strains of live bacteria (450 billion bacteria per sachet) including S. thermophilus DSM24731/NCIMB 30438, Bifidobacterium breve DSM24732/NCIMB 30441, Bifidobacterium longum DSM24736/NCIMB 30435, Bifidobacterium infantis DSM24737/NCIMB 30436, Lactobacillus acidophilus DSM24735/NCIMB 30442, Lactobacillus plantarum DSM24730/NCIMB 30437, Lactobacillus paracasei DSM24733/NCIMB 30439, Lactobacillus delbrueckii ssp. bulgaricus DSM24734/NCIMB 30440 . 64 From lines of affidavits, we have cross-evaluated and found that seven of the strains affect the bilirubin metabolism. It can be stored at room temperature for up to week without having a major effect on potency. Otherwise, it is safe at 4°C–8℃. 65 66 We prepared an icebag that can keep the product at a low temperature during transportation to home and each participant will store the product in a refrigerator at home. It will be stored, distributed and administered as per the product instruction by clinicians.

  • View inline

Summary of probiotic species that have direct or indirect effect on neonatal jaundice from different literatures

Outcome measures

Primary outcome.

Hyperbilirubinaemia will be the primary outcome of this study. For all participants’ babies, sternal transcutaneous bilirubin (TcB) level will be measured within 2 days and on the seventh day of life by Dräger Meter JM-105, which is a non-invasive device. When TcB exceeds or is within 3 mg/dL of the phototherapy treatment threshold or if the TcB is 15 mg/dL, TSB will be measured as a confirmatory test. 67 Hyperbilirubinaemia will be declared when TSB is >5 mg/dL/24 hours or >12 mg/dL after 1 day. Since our intervention will be initiated during pregnancy, all outcomes of each type of jaundice including jaundice diagnosed within 24 hours will be compared between the two groups by considering the intrauterine exposure to probiotics. The TcB measurement device is studied in different settings including a study conducted in Hong Kong in 2016 at Baby-friendly Hospital 68 and Tuen Mun Hospital, 69 and in a tertiary hospital in Malaysia. 70 It is a very quick, effective and non-invasive method of measurement. It is also a reliable method with a better correlation coefficient with TSB measurement. 71–73 The bilirubin level will be plotted on the standard neonatal bilirubin level nomogram chart to decide if the infant has considerable jaundice. A measurement of TcB level above the 95th percentile on the nomogram will be regarded as a high risk for subsequent hyperbilirubinaemia 67 and will be investigated with TSB and followed. 74 There is also a phototherapy and exchange transfusion threshold determining nomogram based on gestational age and age in hours. 67 This threshold-determining nomogram will be used and the need for phototherapy and exchange transfusion will also be assessed as outcomes. By taking in utero exposure into consideration, all newborns irrespective of their health status and admission status to the special care baby unit will be included in the study.

Secondary outcomes

Secondary outcomes involve (1) Pregnancy outcomes will be collected by a questionnaire which contains different outcome measure variables including fetal well-being, preterm labour, pre-eclampsia, diabetes mellitus, infection/sepsis, induction of labour, mode of delivery, GA at birth, birth weight, Apgar score and …etc. (2) Milk and stool microbiome: Breast milk samples will be collected within the second and on the seventh day of giving birth. It will be kept at 4°C during transportation and stored at 80°C freezer. The maternal stool will be collected before initiation of the intervention (at 36 weeks of gestation) and after 3 weeks of intervention. Infant stool will be collected simultaneously with breast milk samples ( figure 2 ). Both maternal and infant stool samples will be collected using a tube containing DNA stabiliser and then aliquoted and frozen at 80°C. Both samples will be processed based on the commercially available kit protocol. Metagenomic sequencing will be used to characterise the microbiome in each sample. For analytical measurement, the number of participants from whom provided probiotics were detected (relative risk) and the mean concentration of provided probiotic species in the breast milk and infant stool will be measured. Maternal stool sample microbiome profiles before and after intervention in both probiotic and placebo groups will be compared. (3) Safety for the mother and fetus/ infant: An independent safety monitoring committee and all research team members will monitor each participant for any adverse events, in particular, gastrointestinal symptoms and possible allergies. Mothers and infants will be followed regularly for possible side effects such as allergic reactions, stomach upset, diarrhoea, flatulence (passing gas) and bloating. 37 Apart from discomfort causing symptoms such as bloating other severe side effects are uncommon. However, in any case, such a moderate to severe side effects, such as decreased/absent/fetal movement, fever, uncommon vaginal fluid leakage, severe headache, decreased abdominal girth or overdistended abdomen, the intervention will be stopped, the participant will be linked to the Accident and Emergency Department. If any adverse events occur in infants, infants will be linked to the paediatrics department.

Timeline of the POPS study from recruitment to end of follow-up. POPS, Perinatal Oral Probiotics Supplementation

Follow-up, compliance and dietary intake data

Pregnant women will be reminded to collect preintervention and postintervention stool samples, start the intervention at 36 weeks of gestation and be advised to report when labour starts or any discomfort. For better retention of participants, a reminder message will be sent to each participant using WhatsApp in the local language at four different time points. The pregnant women will be reminded to record daily on the diary form regarding the dose and time they take the supplement, supplemented with any use of prescribed antibiotics or adverse events such as any diarrhoea, hospitalisation, etc. In addition, they will also be reminded to keep their usual diet only and not to take any other probiotic products. The unused supplements report will be collected. Maternal dietary intake and diversity will be evaluated using standard individual dietary diversity measurement guidelines. 75 76

Data and sample collection

For this randomised controlled clinical trial, data will be collected at least at three time points.

Recruitment: At this stage, participants’ characteristics such as age, marital status, educational level and other sociodemographic characteristics, obstetrics and gynaecological characteristics, medical and surgical factors, reproductive history-related factors including family size, behavioural-related factors (smoking or alcohol use), allergies, use of supplements/vitamins/probiotics, prior lactation history, use of standing dose of antibiotics, plan for the place of birth, current pregnancy condition (gestational age, type of pregnancy (singleton), presentation, fetal heartbeat, placental location, fetal weight, any supplement/ vitamin/ probiotic in the previous 1 month. Preintervention maternal stool samples will be collected from women in both groups. The intervention will be initiated at 36 weeks of gestation and after 3 weeks of intervention, postintervention data will be collected.

Within 2 days of life and before discharge:

Having sufficient rest after delivery, mothers will be contacted until before discharge home for data collection at their most convenient time. The time for data collection will be marked. Data including gestational age at delivery, date and time of delivery, mode of delivery, any antibiotic use; infant birth weight, Apgar score, urination status of the infant and time of initiation of breast feeding will be documented. Breast milk and infant stool samples will be collected, and the volume of samples collected, and storage temperature for both samples. Moreover, the neonate’s TcB level will be measured and documented.

Follow-up on the seventh day of infant life: Breast milk and infant stool samples will be collected. Both the volume and storage temperature of samples will also be recorded. Infant body weight and bilirubin level will be measured. Breastfeeding status will be recorded. The diary record for supplements will be returned and the final sachet count report will be recorded.

In general, the data collection will be exclusively by the research staff appointed for this research using a pretested questionnaire. After recruitment, every mother will be counselled at each contact for better compliance with the intervention.

Data handling and quality assurance

All the data collected through this study will be handled by the principal investigator and researchers and kept confidential. Data quality will be checked regularly by all investigators separately and a trial managing committee.

Sample size

The sample size in this protocol is calculated using the G*power V.3.1.9.4 statistical software. 77 Due to the lack of previous studies on pregnant women targeting bilirubin level reduction in neonates, we used a previous study conducted to assess the effect of a probiotic on neonatal hyperbilirubinaemia by providing the lactobacillus RGG directly to neonates in Turkey to calculate our study sample size (PMID: 30968632). 78 The effect size was calculated using mean, SD and sample size, which are the most common parameters to measure effect size. In the paper conducted in Turkey, the mean and SD of bilirubin level and the sample size were mentioned, and we took the third-day bilirubin measurement records. The effect size was computed as 0.663. Finally, the sample size is calculated using the following assumptions: Two tailed, power 80, effect size 0.663, α=0.05, 1:1 allocation ratio. This yields about 74 (37 in each arm), and then we considered a 20% drop-out using the formula N=(n/1−DO). The final sample size, therefore, computed 94 (47 in each group). 48 We have also calculated the sample size using a reduction of the incidence of neonatal jaundice in controls (p=46.6%) and in the probiotics group (p=14.6%) and yield (n=62; 31 in each group), which is smaller than the first sample size. To calculate this sample size, we used additional parameters (α=0.05, power=0.8, enrolment ratio 1:1) and calculated using an online platform. 79

Statistical analysis

This study will use the Consolidated Standards of Reporting Trials guidelines. 80 An intention-to-treat approach will be used to assess the cause-and-effect relation to maintain the effect of randomisation. Baseline characteristics will be compared between allocated treatment groups using frequencies and descriptive statistics. Pearson’s χ 2 test and Fisher’s exact test will be run to assess the difference of different factors between the two intervention groups ( online supplemental table ). To assess the exclusive effect of the intervention on the occurrence of neonatal jaundice, a binary logistic regression model will be fitted. Additionally, to examine the effect of the intervention on the mean bilirubin level of neonates, mean beneficial bacteria level in breast milk, and in maternal and infant stool, linear regression will be employed. The microbial DNA will be extracted from different specimens, including breast milk, and maternal and infant stool samples using commercial kits. Quality control and metagenomics sequencing will be employed and analysed by bioinformatics experts to obtain microbiome information including taxonomy and function.

Study status

Participant recruitment has not started yet.

Patient and public involvement

The public representative was involved in the Joint CUHK-NTEC Clinical Research Ethics Committee, whose membership includes lay persons from the community, apart from clinical and nursing staffs. Results of the trial will be disseminated to the community including study participants through presentations in workshops and posters as well as flyers that will be posted in the outpatient clinics for ease of access. Plan for participant involvement in participant recruitment is not considered in this study. This trial has subsequent follow-up schedules. In each visit, participants will be asked for any adverse effects. A diary form is also prepared to mark down the dose taken and any side effects.

Ethics and dissemination

All the procedures in this clinical trial will be conducted as per the ethical principles of the Declaration of Helsinki of the World Medical Association and the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) Guideline for Good Clinical Practice (GCP) for medical research involving human subjects. Ethical approval was obtained from the CUHK-NTEC (The Chinese University of Hong Kong and New Territories East Cluster) Clinical Research Ethics Committee (CREC) Hong Kong (with CREC Reference number 2023.100-T). Participants will be identified and screened with eligibility criteria by reviewing their medical records and questioning them for these criteria. For those who fulfilled the inclusion criteria, adequate information will be provided by using the printed information sheet. Written informed consent will be taken from each participant after their approval. Codes instead of names will be used and all data will be kept confidential. These codes will be used throughout each communication among the research team and for data analysis.

In focus on increasing visibility, using any recognised benefits of the interventions resulting from this study and further evaluation, enhancing academic collaborations, linking relevant research areas in this field and identifying further areas where research could be effectively managed, this study result will be disseminated through presentations in academic seminars, workshops, and national and international symposiums. It will be submitted to The Chinese University of Hong Kong for archival and possible dissemination. The aggregated findings of this trial will be published in peer-reviewed academic journals for better visibility. A lay summary of the results and links to publications will be made available on databases.

Emerging evidence increasingly indicates that the microbiome established in early life, even before birth, plays a pivotal role in shaping human health and disease outcomes in later years. 81 Neonatal jaundice, a prevalent problem in newborns, is attributed to dysfunction in bilirubin metabolism, production and excretion. 82 Despite treatments such as phototherapy and exchange transfusion are commonly employed, however, preventive approaches for neonatal jaundice are limited. Gut flora is instrumental in converting conjugated bilirubin into stercobilinogen and urobilinogen, facilitating its excretion as stercobilin in faeces and urobilin in urine. 20 Interestingly, the neonatal gut microbiome is primarily established by the maternal gut and milk microbiome during pregnancy and lactation periods. 83 84 Therefore, remodelling the maternal microbiome provides us with a potential opportunity to shape the gut microbiome in newborns.

Probiotics have been proven to be an effective intervention to shape the maternal-offspring microbiome. Of note, lines of evidence suggest maternal probiotic supplementation not only positively influences the breast milk and infant’s gut microbiome but also enhances maternal health during pregnancy. 26 Importantly, no adverse effects from these supplements during pregnancy have been reported to date. Previous studies showed that directly administering probiotics to neonates enables them to treat and prevent neonatal jaundice. 39 85 However, direct neonatal administration of probiotics is not always practical. Therefore, we questioned if breast milk can be modulated by probiotic supplementation during pregnancy thereby reshaping the infant’s gut microbiome and reducing neonatal complications such as jaundice.

To this end, we hypothesised perinatal probiotic supplementation in pregnancies may serve as a feasible approach to preventing neonatal jaundice and designed this randomised controlled trial to evaluate its effect. Our study will revolutionise current neonatal care practices, offering a non-invasive, safe, inexpensive and potentially highly effective preventive strategy for neonatal jaundice. It also underscores the critical role of the maternal microbiome in infant health, paving the way for new approaches in prenatal care.

Ethics statements

Patient consent for publication.

Consent obtained directly from patient(s).

Acknowledgments

The Hong Kong Obstetrical & Gynaecological Trust Fund granted funding for the team to handle this study. The Clinical Research Pharmacy in the Prince of Wales Hospital, Hong Kong has signed agreement with this study team to store the investigational food products (Vivomixx and placebo) and dispense by keeping the cold chain of the product. The authors would like to acknowledge Mendes, the innovative microbiotic company in Europe, for supplying the investigational food product (Vivomixx and placebo). The trial team would like to sincerely acknowledge the Department of Obstetrics and Gynaecology, at the Chinese University of Hong Kong for supporting the trial in logistics.

  • Maisels MJ ,
  • McDonagh AF
  • Burman-Roy S ,
  • Murphy MS , et al
  • Olusanya BO ,
  • Kandasamy D ,
  • Roginski C , et al
  • Osibanjo FB ,
  • Tiwari PK ,
  • Kumar A , et al
  • Hsiao K-J ,
  • Wuu K-D , et al
  • Li Z-K , et al
  • Trasancos C ,
  • Li A , et al
  • Coleman K , et al
  • Villaveces A ,
  • Dhillon P , et al
  • Wong RM , et al
  • Agarwal R ,
  • Akagawa S ,
  • Akagawa Y ,
  • Yamanouchi S , et al
  • Dufault-Thompson K , et al
  • Li J , et al
  • Zhang W , et al
  • Wu L , et al
  • Rescigno M ,
  • Valzasina B , et al
  • Macpherson AJ ,
  • Day AS , et al
  • Turroni F ,
  • Ventura M , et al
  • Moubareck CA
  • Virmani MD ,
  • Rosa F , et al
  • Cookson AL ,
  • Otter DE , et al
  • Santosa I ,
  • Itoh S , et al
  • Williams NT
  • Korpela K ,
  • Salonen A ,
  • Vepsäläinen O , et al
  • Wang T , et al
  • Duman N , et al
  • Kirpich IA ,
  • Solovieva NV ,
  • Leikhter SN , et al
  • He F , et al
  • Kulkarni N ,
  • De Preter V ,
  • Cloetens L , et al
  • Torkaman M ,
  • Mottaghizadeh F ,
  • Khosravi MH , et al
  • Quigley EMM
  • O’Hara AM ,
  • O’Regan P ,
  • Fanning A , et al
  • Aslan Y , et al
  • Aldemir M ,
  • Hosoglu S , et al
  • Collado MC ,
  • Jalonen L ,
  • Meriluoto J , et al
  • Aiyegoro OA ,
  • Okpeku M , et al
  • Sahariah SA ,
  • Chopra H , et al
  • Purdue-Smithe AC ,
  • Schliep KC , et al
  • Cruciani F ,
  • Baldassarre ME , et al
  • Facchinetti F ,
  • Pedretti L , et al
  • Mastromarino P ,
  • Capobianco D ,
  • Miccheli A , et al
  • Jafarnejad S ,
  • Jafarnejad F , et al
  • Baldassarre ME ,
  • Di Mauro A ,
  • Mastromarino P , et al
  • Sarangi AN ,
  • Goel A , et al
  • Astolfi ML ,
  • Protano C ,
  • Schiavi E , et al
  • Halkjær SI ,
  • de Knegt VE ,
  • Lo B , et al
  • Tetzlaff Jm Fau - Altman DG ,
  • Altman Dg Fau - Laupacis A , et al
  • De-Simone-Formulation
  • Atukunda EC ,
  • Owembabazi M ,
  • Pratt MC , et al
  • Mintjens S ,
  • van Poppel MNM ,
  • Groen H , et al
  • McCarthy JS ,
  • Yalkinoglu Ö ,
  • Odedra A , et al
  • Kemper AR ,
  • Newman TB ,
  • Slaughter JL , et al
  • Chiu-Fan LAU ,
  • Mohamed M ,
  • Ibrahim NR ,
  • Ramli N , et al
  • Vandermeer B ,
  • Campbell S , et al
  • Hassan Shabuj M ,
  • Hossain J ,
  • Chenarani RM ,
  • Maamouri G ,
  • Khodashenas E , et al
  • Kennedy G ,
  • Ballard T ,
  • Ganpule-Rao AV ,
  • Yajnik CS , et al
  • Buchner A ,
  • Erdfelder E ,
  • Faul F , et al
  • Sereika SM ,
  • ↵ Sample Size Calculator: Determines the minimum number of subjects for adequate study power . Updated July 24, 2019. Accessed June 4, 2024. Available : https://clincalc.com/stats/samplesize.aspx
  • Waechter R ,
  • Burgen KS ,
  • Punch B , et al
  • Stiemsma LT ,
  • Woodgate P ,
  • Mueller NT ,
  • Combellick J , et al
  • Zheng T , et al
  • Palladino V ,
  • Amoruso A , et al
  • Kumral A Fau - Duman A ,
  • Fau - Duman N , et al
  • Sun Q , et al

Supplementary materials

Supplementary data.

This web only file has been produced by the BMJ Publishing Group from an electronic file supplied by the author(s) and has not been edited for content.

  • Data supplement 1
  • Data supplement 2

Contributors SLL is the principal investigator of this trial. CCW and BKA conceived the idea. BKA developed the research question and study design and prepared the protocol for ethical approval submission under the supervision of CCW and YW. WFL and BKA prepared an information sheet, consent form and other logistics before submission to the ethics committee for approval. MBWL and BKA optimised the laboratory sample handling, storage and processing protocol. BKA, CCW and YW established collaboration with the innovation microbiotic company for free supply of the investigational food product. MWL, SLL, CCW, YW, BKA and WFL obtained the funding. CCW is the senior trial supervisor who coordinated the operational delivery of the study protocol. WFL provides support on recruitment. YW and BKA handle the data and analysis. BKA drafted the manuscript and all authors listed provided critical review and approved the manuscript for submission.

Funding The Hong Kong Obstetrical & Gynaecological Trust Fund supported the trial (grant number: N/A). Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong provided logistics for the laboratory sample storage, processing and analysis.

Competing interests None declared.

Patient and public involvement Patients and/or the public were not involved in the design, or conduct, or reporting, or dissemination plans of this research.

Provenance and peer review Not commissioned; externally peer reviewed.

Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.

Read the full text or download the PDF:

  • Type 2 Diabetes
  • Heart Disease
  • Digestive Health
  • Multiple Sclerosis
  • Diet & Nutrition
  • Supplements
  • Health Insurance
  • Public Health
  • Patient Rights
  • Caregivers & Loved Ones
  • End of Life Concerns
  • Health News
  • Thyroid Test Analyzer
  • Doctor Discussion Guides
  • Hemoglobin A1c Test Analyzer
  • Lipid Test Analyzer
  • Complete Blood Count (CBC) Analyzer
  • What to Buy
  • Editorial Process
  • Meet Our Medical Expert Board

Jaundice: Everything You Need to Know

More Than Just Yellow Skin and Eyes

  • Complications
  • When to Seek Care

Jaundice , also known as icterus , is the yellowish discoloration of the skin and eyes caused by the abnormal buildup of an orangish waste product called bilirubin . Bilirubin is produced by the normal breakdown of red blood cells (RBCs) and processed by the liver , where it is cleared from the body in bile . If too much bilirubin is produced or the liver cannot clear it, jaundice can develop.

Jaundice is not dangerous, but the underlying cause can be. Causes range from relatively benign conditions like Gilbert syndrome to potentially fatal ones like liver cancer . Jaundice can occur at any stage of life, including birth ( neonatal jaundice ). The treatment varies by the underlying cause.

This article describes the causes, symptoms, and diagnosis of jaundice in adults and children. It also explains how jaundice is treated and prevented, including when jaundice is a sign of a medical emergency.

This photo contains content that some people may find graphic or disturbing.

ZayWin Htal / Getty Images

Types and Causes of Jaundice

Jaundice occurs when there is too much bilirubin in the body, referred to as hyperbilirubinemia . There are three main reasons why this might occur, which are:

  • Prehepatic jaundice : Too many red blood cells are being broken down, and the liver is unable to handle the overload, causing bilirubin to escape into the bloodstream. ("Hepatic" refers to the liver.)
  • Hepatic jaundice : The liver is damaged and doesn't remove enough bilirubin from your blood.
  • Posthepatic jaundice : There is an obstruction in the  biliary system (composed of the liver, gallbladder , bile ducts, and pancreas ) that causes bile to back up into your liver.

The other classification of neonatal jaundice is an otherwise normal occurrence in newborns but one that can turn serious in some.

Prehepatic Jaundice

Red blood cells have a life span of around 120 days. They are broken down by the body and replaced by new ones. The process, called hemolysis , produces around 4 milligrams (mg) of unconjugated (free-circulating) bilirubin per day, which the liver usually disposes of in urine and stool.

Prehepatic jaundice is caused when hemolysis is increased, overwhelming the liver with more bilirubin than it can handle.

Possible causes of this include:

  • Hemolytic anemia (low numbers of RBCs due to increased destruction)
  • Sickle-cell anemia (a hereditary condition of variant hemoglobin-the oxygen-carrying molecule in RBCs)
  • Thalassemia (an inherited condition affecting hemoglobin production)
  • Spherocytosis (having spherical RBCs rather than biconcave ones)
  • Alpha-1 antitrypsin (A1AT) deficiency (an inherited disorder affecting a liver enzyme, a substance that affects chemical reactions)
  • Glucose-6-phosphate dehydrogenase (G6PD) deficiency (an inherited disorder affecting an enzyme in RBCs)
  • Severe malaria infection (a mosquito-borne parasitic infection affecting RBCs)

Hepatic Jaundice

Once unconjugated bilirubin enters the liver, it is exposed to enzymes that transform it into conjugated bilirubin. This form can be incorporated into a digestive fluid called bile and eliminated from the body as it is carried away in the stool.

Hepatic jaundice is caused when liver cells involved in this process, called hepatocytes , are damaged. The damage may be transient (temporary) or permanent, caused by a wide range of infectious, autoimmune, inflammatory, and genetic diseases like:

  • Viral hepatitis (most commonly hepatitis A , hepatitis B , and hepatitis C )
  • Alcoholic liver disease
  • Metabolic dysfunction-associated steatotic liver disease (MASLD) (fat accumulation and inflammation in the liver)
  • Liver cirrhosis (extensive scarring in the liver)
  • Autoimmune hepatitis
  • Drug-induced liver toxicity
  • Hemochromatosis (iron overload)
  • Wilson’s disease (genetic disorder leading to a buildup of copper)
  • Gilbert syndrome (a genetic condition affecting a liver enzyme)
  • Liver cancer

Posthepatic Jaundice

Once bile leaves the liver, it is transported to the gallbladder for storage. Whenever food is eaten, it is released into the main duct, called the common duct, where it mixes with digestive fluids from the pancreas. These fluids are then released into the first part of the small intestine , called the duodenum , to break down fats and protein.

Posthepatic jaundice occurs when there is an obstruction in the pathway from the liver to the small intestine. This causes bile to back up into the liver, where bilirubin can escape.

Posthepatic jaundice may be due to an obstruction or a disease that causes bile ducts to become narrow or pinched. Examples include:

  • Biliary tract strictures (narrowing of bile ducts)
  • Primary biliary cirrhosis (damage to the small bile ducts due to inflammation)
  • Primary sclerosing cholangitis (damage to the small bile ducts due to inflammation)
  • Pancreatitis (inflammation of the pancreas)
  • Cholestasis of pregnancy (slowing of bile flow)
  • Liver flukes (parasitic infection)
  • Gallbladder cancer
  • Bile duct cancer
  • Pancreatic cancer

Neonatal Jaundice

Neonatal jaundice is a common occurrence in newborns, affecting 60% of full-term babies and 80% of preterm babies . It usually develops by the second or third day of life and almost always clears on its own without consequence.

Neonatal jaundice is caused when fetal hemoglobin (the protein in red blood cells that carries oxygen) is broken down and replaced with the post-birth form of hemoglobin. This breakdown causes the release of more bilirubin than a newborn's liver can clear.

On rare occasions, neonatal jaundice is not normal. Some of the same conditions that cause jaundice in adults—like G6PD deficiency, spherocytosis, and A1AT deficiency—can also affect newborns, leading to potentially life-threatening complications,

Other serious causes in newborns include:

  • Biliary atresia (blocked bile ducts)
  • Alagille syndrome (a genetic condition affecting bile flow)
  • Rh factor hemolytic disease (a blood type mismatch between the pregnant person and the fetus)
  • Familial cholestasis (a genetic condition in which bile builds up in liver cells)

Justin Paget / Getty Images

What Is Breastfeeding Jaundice?

Another relatively benign cause of jaundice is "breastfeeding jaundice," a casual term for suboptimal intake jaundice. This usually occurs in the second week of life when a newborn is not getting enough nutrition.

Inadequate feeding delays the passage of the baby's first poop (called the meconium ), which contains large amounts of bilirubin. Breast milk is also thought to contain substances that can impair the conjugation of bilirubin in the liver.

Increased breastfeeding or supplementation with bottled formula will almost invariably help resolve the condition.

Jaundice Symptoms

Jaundice is characterized by the yellowing of the skin and sclera (white of the eye) due to the buildup of bilirubin into tissues. Depending on the cause, the symptoms can be transient and barely noticeable or long-lasting and severe.

Jaundice symptoms can also differ in newborns.

Reproduced with permission from ©DermNet and ©Te Whatu Ora dermnetnz.org 2023.

Common Symptoms

On its own, jaundice does not cause anything other than yellowish discoloration. In many cases, it is the first sign of a disease and, in some, the only sign.

Jaundice in adults and children is typically pathologic, meaning it is related to a disease. If other symptoms accompany jaundice, it is due to the underlying disease. These symptoms can vary by whether the condition is hepatic, posthepatic, or prehepatic.

Hepatic and posthepatic causes of jaundice often manifest with symptoms of hepatitis (liver inflammation), causing:

  • Nausea and vomiting
  • Upper-right abdominal pain and swelling
  • Dark urine (caused by the buildup of bilirubin in your urine)
  • Pale and greasy stool (caused by the lack of bile secretion)

Prehepatic jaundice caused by excessive hemolysis can lead to symptoms of hemolytic anemia , causing:

  • Rapid heart rate
  • Shortness of breath
  • Low exercise tolerance
  • Dizziness or light-headedness
  • Enlarged liver or spleen
  • Blood in urine

Symptoms in Newborns

Neonatal jaundice is most often physiological , meaning it is related to normal bodily functions. Symptoms tend to develop within two to three days of birth, starting at the face and moving downward to the chest, stomach, legs, and feet.

Depending on the severity, there may be other symptoms such as excessive sleepiness, fussiness, and poor feeding, Over time, though, the yellowing will usually dissipate as large amounts of bilirubin are excreted in the baby's stool and urine.

However, in some newborns, jaundice may be pathologic. This is typically the case when bilirubin levels are extremely high or when jaundice occurs immediately after birth or persists for weeks.

Persistent hyperbilirubinemia can lead to a potentially fatal condition called kernicterus , in which bilirubin invades the brain, causing brain dysfunction known as encephalopathy . If not treated immediately, kernicterus can lead to seizures, brain damage, and complications such as:

  • Involuntary eye movements ( nystagmus )
  • Uncontrollable twitches and spasms ( myoclonus )
  • Permanent hearing loss
  • Learning disabilities
  • Cerebral palsy (a neurological condition due to brain damage)

How Long Does Jaundice Last?

The duration of jaundice varies based largely on whether the underlying condition is acute (sudden and severe) or chronic (persistent or long-lasting). Generally speaking, the discoloration will fade once the underlying cause is resolved—whether it be an acute infection, biliary obstruction, or toxic drug reaction.

Jaundice has a typical course with some diseases (like acute viral hepatitis , which usually peaks in two weeks and starts to fade over two to three weeks). With other diseases, such as liver cancer, jaundice will only clear after a liver transplant .

With neonatal jaundice, the timeline is more consistent. In formula-fed babies, jaundice typically clears within two weeks. In breastfed babies, jaundice may last two to three weeks, sometimes longer.

How Jaundice Is Diagnosed

While jaundice is usually self-evident, a physical exam and lab tests are needed to help narrow the possible causes.

As part of the evaluation, the healthcare provider will perform a urinalysis to check for bilirubin in your urine and liver function tests (LFTs) to measure levels of bilirubin and another byproduct called urobilinogen in your blood.

LFTs also measure liver enzymes called alkaline phosphatase (ALP) , alanine transferase (ALT) , and aspartate transferase (AST), which tend to rise with liver or biliary disease.

These results, paired with the color of your urine and stool, can help determine if the cause is prehepatic, hepatic, or posthepatic.

  Prehepatic  Hepatic  Posthepatic
Total bilirubin Normal or high High High
Unconjugated bilirubin Normal High Normal
Conjugated bilirubin Normal High High
Bilirubin in urine Not present Present Present
Urine color Normal Dark Dark
Stool color Brown Pale Pale to white
ALT and AST Normal Very high High
ALP Normal High Very high
Urobilirubin Normal to high Low Low to none

Other tests and procedures may be ordered based on the initial findings, including imaging studies like ultrasound , computed tomography (CT) , or magnetic resonance imaging (MRI) to check for blockages. A liver biopsy may also be performed to obtain a sample of liver tissue for evaluation in the lab.

Neonatal jaundice is monitored with blood tests that measure bilirubin in milligrams per deciliter of blood (mg/dL). Bilirubin can also be measured with a noninvasive, handheld device called a transcutaneous bilirubinometer , though blood testing remains the gold standard.

With physiological jaundice, newborns typically have unconjugated bilirubin levels between 12 and 15 mg/dL for the first two weeks, gradually decreasing to 2 mg/dL over the next month and eventually settling to a normal adult level of 0.2 to 0.8 mg/dL.

On the other hand, jaundice is considered pathologic if:

  • Total bilirubins are over 19.5 mg/dL.
  • Total bilirubins increase by 5 mg/dL per day or 0.5 mg/dL per hour.
  • Jaundice occurs within the first 24 hours of birth.
  • Jaundice persists without letting up for more than two weeks.

Jaundice Treatment

Jaundice is treated by resolving the underlying condition. The treatment varies by the cause.

Treatment may be provided by different specialists, such as:

  • Gastroenterologist (digestive system specialist)
  • Hepatologist (liver specialist)
  • Hematologist (blood disorder specialist)
  • Oncologist (cancer specialist)
  • Rheumatologist (autoimmune, inflammatory, and musculoskeletal disease specialist)
  • Surgical team

If a bile duct is blocked, surgery or procedures like shock wave lithotripsy can help clear the obstruction. Surgery may also be needed for cancer of the liver, pancreas, gallbladder, or bile duct, along with chemotherapy and radiation .

When a chronic disease like cirrhosis or primary sclerosing cholangitis causes liver failure (meaning that the liver can no longer support the body's needs), the only option may be a liver transplant .

In all other cases, the treatment is largely medical.

While most cases of jaundice in newborns resolve on their own, those with total bilirubins over 21 mg/dL should receive specialized phototherapy (sometimes referred to as "bili lights").

Phototherapy involves exposure to a certain bandwidth of blue light (not ultraviolet light) that converts bilirubin in the skin into a water-soluble form that is more easily passed in urine. Home phototherapy machines allow home treatment outside of the hospital.

In severe cases, a procedure called an exchange transfusion can counteract the effects of kernicterus. Exchange transfusion is performed by slowly removing blood from the infant and replacing it with fresh donor blood or plasma. While potentially lifesaving, risks include blood clots, severe changes in blood chemistry, and shock .

How to Prevent Jaundice

Jaundice can't always be prevented, but there are certain measures you can take to avoid some of the underlying causes, including:

  • Eat a healthy, balanced diet low in fat and high in vegetables, fruits, and whole grains. This can reduce your risk of MASLD, gallstones, and acute pancreatitis.
  • Limit your alcohol intake to no more than two drinks per day for males and one day per drink per day for females. If you already have liver disease, alcohol may need to be avoided.
  • Get vaccinated against hepatitis A and hepatitis B .
  • Quit smoking . Smoking causes the constriction (narrowing) of blood vessels that can contribute to liver and biliary disease.
  • Avoid the overuse of Tylenol (acetaminophen) , which is associated with a high risk of hepatotoxicity (liver poisoning). Never take Tylenol with alcohol.
  • Avoid herbal remedies like kava, ephedra, skullcap, and pennyroyal, all of which have been linked to acute liver failure.
  • To reduce the risk of breastfeeding jaundice, newborns should have eight to 12 feedings per day for the first several days of life. Formula-fed infants should have 1 to 2 ounces (30 to 60 milliliters) of formula every two to three hours for the first week.

Complications of Jaundice

Left untreated, the underlying causes of jaundice can turn serious and even life-threatening.

One of the key concerns is acute liver failure—also known as fulminant hepatic failure —which can sometimes develop rapidly in otherwise healthy individuals. This is especially true with severe drug-induced liver injury, in which a single event can cause irreparable damage requiring a liver transplant.

Autoimmune hepatitis and acute hepatitis A and B have also been known to cause acute liver failure.

Another concern is complications of hemolytic anemia, one of the most common causes of prehepatic jaundice. Severe cases have been known to cause life-threatening arrhythmia (irregular heartbeats), heart failure , and even death.

When to Contact a Healthcare Provider

No amount of jaundice is considered "normal." Call a healthcare provider immediately if you develop jaundice for any reason—even if it is mild and you have no other symptoms. Early intervention can help you avoid potentially serious complications.

Seek immediate emergency care if jaundice is accompanied by:

  • Sudden, severe sleepiness
  • A fruity breath
  • Disorientation or confusion
  • A swollen, fluid-filled belly ( ascites )

When to Call 911: Neonatal Jaundice

Call 911 or rush to the nearest emergency department if your baby has jaundice accompanied by signs of kernicterus, such as:

  • A shrill, high-pitched cry
  • Extreme drowsiness
  • Poor feeding
  • Head and limb loppiness, like a rag doll
  • The lack of a startle reflex
  • Gaps in breathing

Jaundice is the yellowing of the skin and eyes caused by the abnormal buildup of bilirubin. Bilirubin is a byproduct of the breakdown of red blood cells.

Jaundice can be prehepatic (related to the excessive breakdown of red blood cells), hepatic (related to the liver), or posthepatic (related to the impaired flow of bile from the liver). Neonatal jaundice in newborns is usually normal but can become life-threatening if bilirubin levels are persistently high.

Jaundice is diagnosed with blood tests and will usually clear once the underlying condition is resolved. Infants with very high bilirubin levels may benefit from phototherapy or an exchange transfusion.

MedlinePlus.  Jaundice .

Fargo MV, Grogan SP, Saguil A. Evaluation of jaundice in adults . Am Fam Physician. 2017;95(3):164-168.

National Institute of Health and Care Excellence. Jaundice in babies born under 28 days .

Markovic AP, Lalosevic MS, Mijac DD, et al. Jaundice as a diagnostic and therapeutic problem: a general practitioner’s approach . Dig Dis. 2020;40(3):362–369. doi:10.1159/000517301

Thiagarajan P, Parker CJ, Prchal JT. How do red blood cells die? Front Physiol. 2021;12:655393. doi:10.3389/fphys.2021.655393

Gondal B, Aronsohn A. A systematic approach to patients with jaundice . Semin Intervent Radiol. 2016;33(4):253–258. doi:10.1055/s-0036-1592331

Boyer JL, Soroka CJ. Bile formation and secretion: an update . J Hepatol. 2021;75(1):190-201. doi:10.1016/j.jhep.2021.02.011

Eghbalian F, Hasanpour-Dehkordi A, Raeisi R.  The effects of clofibrate on neonatal jaundice: a systematic review .  Int J Prev Med . 2022;13(3). doi:10.4103/ijpvm.IJPVM_407_20

Ullah S, Rahman K, Hedayat M. Hyperbilirubinemia in neonates: types, causes, clinical examinations, preventive measures and treatments: a narrative review article . Iran J Public Health. 2016;45(5):558–568.

Centers for Disease Control and Prevention. Breastfeeding: jaundice .

American College of Surgeons Division of Education. Jaundice .

Mitra S, Rennie J.  Neonatal jaundice: aetiology, diagnosis, and treatment .  Br J Hosp Med (Lond) . 2017;78(12):699-704. doi:10.12968/hmed.2017.78.12.699

National Institute of Diabetes and Digestive and Kidney Diseases.  Hepatitis A .

Pan JJ, Fontana RJ.  CAQ corner: acute liver failure management and liver transplantation .  Liver Transpl.  2022;28(10):1664–1673. doi:10.1002/lt.26503

March of Dimes. Newborn jaundice .

Kemper AR, Newman TB, Slaughter JL. Clinical practice guideline revision: management of hyperbilirubinemia in the newborn infant 35 or more weeks of gestation . Pediatrics. 2022;150(3):e2022058859. doi:10.1542/peds.2022-058859

Wang L, Yu WF. Obstructive jaundice and perioperative management .  Acta Anaesthesiologica Taiwanica . 2014;52(1):22–29. doi:10.1016/j.aat.2014.03.002

Tariq NUA, McNamara MG, Valle JW. Biliary tract cancers: current knowledge, clinical candidates and future challenges . Cancer Manag Res. 2019;11:2623–2642. doi:10.2147/CMAR.S157092

American Liver Foundation.  Liver transplantation .

Storck LJ, Imoberdorf B, Ballmer PE. Nutrition in gastrointestinal disease: liver, pancreatic, and inflammatory bowel disease . J Clin Med. 2019;8(8):1098. doi:10.3390/jcm8081098

National Institute on Alcohol Abuse and Alcoholism. Drinking levels defined .

National Cancer Institute. New insights into why smoking causes fatty liver disease .

National Institute of Diabetes and Digestive and Kidney Diseases.  Clinical and research information on drug-induced liver injury: acetaminophen .

National Institute of Diabetes and Digestive and Kidney Diseases. Herbal and dietary supplements .

Wendon J, Cordoba J, Dhawan A, et al.  EASL clinical practical guidelines on the management of acute (fulminant) liver failure .  J Hepatol . 2017;66(5):1047-1081. doi:10.1016/j.jhep.2016.12.003

National Heart, Lung and Blood Institute.  Hemolytic anemia .

By James Myhre & Dennis Sifris, MD Dr. Sifris is an HIV specialist and Medical Director of LifeSense Disease Management. Myhre is a journalist and HIV educator.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Paediatr Child Health
  • v.11(10); 2006 Dec

Logo of pchealth

Case 2: Newborn with jaundice and “hyperglycemia”

A nine-day-old boy was admitted to the paediatric ward of a regional hospital after a maternal-newborn clinic appointment, where he was found to be jaundiced with a total serum bilirubin of 400 μmol/L. He had been followed by the public health nurse in the community for poor weight gain in his first week of life. His weight on the day of admission was 2.52 kg, while his birth weight had been 2.81 kg.

The pregnancy and family history were unremarkable, and the boy was born at term with an uncomplicated initial stay in the hospital. Breastfeeding had been established before discharge home on the second day of life and was supplemented with formula by bottle after an appointment on the fifth day at the maternal-newborn clinic, where the boy was found to have lost over 10% of his birth weight. His parents reported that he was becoming less and less interested in eating, and by the day of admission, they needed to wake him for most feeds.

On admission, he was jaundiced and moderately dehydrated but otherwise had a normal physical examination. Other than the hyperbilirubinemia, the remainder of the initial blood work was unremarkable. The direct antiglobulin test was negative. The baby was started on double phototherapy and intravenous rehydration, and within 6 h, he had a repeat total bilirubin of 309 μmol/L, with a direct bilirubin of 34 μmol/L. A glucose meter check also performed at the time showed a reading of 26 mmol/L. A serum sample sent to the laboratory showed the glucose to be 2 mmol/L. Repeat glucose meter checks performed overnight were all greater than 20 mmol/L, while laboratory samples were in the low to normal range. After approximately 24 h of phototherapy, the total bilirubin was 250 μmol/L, with a direct bilirubin of 39 μmol/L. The baby developed a bronzed colour (direct hyperbilirubinemia), and liver function tests showed an alkaline phosphatase of 685 U/L, alanine aminotransferase of 83 U/L and aspartate aminotransferase of 155 U/L. A urine test result was able to quickly point to the likely diagnosis.

CASE 2 DIAGNOSIS: GALACTOSEMIA (GALACTOSE-1-PHOSPHATE URIDYL TRANSFERASE DEFICIENCY)

The urine was strongly positive for reducing substances and negative for glucose. Other studies performed as part of the evaluation of the direct hyperbilirubinemia included abdominal ultrasound (to assess liver and biliary anatomy); alpha-1-antitrypsin level; urine for viral culture; thyroid function; metabolic screen; and hepatitis viral serology. A confirmatory red blood cell study of galactose-1-phosphate uridyl transferase (GALT) activity showed markedly reduced quantity. The baby was started on a lactose-free soy formula two days after admission, resulting in a marked improvement in feeding, growth and liver function tests.

The differential diagnosis for a newborn with a direct hyperbilirubinemia (conjugated bilirubin greater than 30 μmol/L or greater than 15% of total bilirubin) is broad. Etiologies can be divided into three general categories ( 1 ): obstruction to biliary flow, hepatic cell injury and chronic bilirubin overload. Obstruction to biliary flow can be secondary to anatomic abnormalities (eg, extrahepatic biliary atresia) or from the production of viscous bile (eg, cystic fibrosis). Hepatic injury can be from numerous sources, such as congenital infections, parenteral nutrition or inborn metabolic errors (eg, galactosemia). Chronic bilirubin overload is less commonly a cause of direct hyperbilirubinemia in the newborn period but can be seen with hemolytic anemias, such as glucose-6-phosphate dehydrogenase (G6PD) deficiency or hereditary spherocytosis.

Initial investigations should confirm a cholestatic picture, establish liver function and detect readily treatable disorders. Secondary investigations should identify a specific diagnosis among the more common etiologies. Preliminary investigations include total and direct bilirubin levels, blood and urine cultures, direct antiglobulin test, alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, gamma-glutamyl transferase, serum glucose, albumin, ammonia, coagulation studies and a complete blood count with differential. To further delineate a diagnosis, more specific testing can be done, such as an abdominal ultrasound, alpha-1-antitrypsin, congenital infection screen, metabolic screen (blood gas, serum amino acids and urine organic acids), sweat chloride, urine for reducing substances (see below), thyroid studies or other tests as indicated.

Galactosemia is an autosomal recessive disorder in which galactose is not properly metabolized. Dietary lactose is broken down by lactase into glucose and galactose and then, in a three-step process, galactose is converted to glucose. This metabolic pathway is particularly important for the newborn, whose main carbohydrate source is lactose. Classic galactosemia, by far the most common variant, involves a deficiency of GALT. This results in a buildup of galactose-1-phosphate and other precursors, causing damage to many organs, including the liver, spleen, kidney, ocular lens, cardiac muscle, brain, gonadal tissue and erythrocytes. The earlier the disorder is diagnosed and a galactose-free diet is implemented, the less damage will ensue. Even with optimal dietary treatment, developmental delays and learning disabilities are common, and affected women almost invariably suffer premature ovarian failure ( 2 ).

Galactosemia has an incidence of approximately one in 60,000 in North America ( 3 ). Several countries have universal newborn screening for galactosemia, but the practice is not widespread. In Canada, only a minority of provinces have a newborn galactosemia screening program in place. More common symptoms and signs suggestive of galactosemia are as follows ( 4 ):

  • poor feeding or weight gain
  • hepatomegaly
  • encephalopathy
  • full fontanel
  • bleeding or easy bruising

There are also several reports of galactosemic infants presenting with Escherichia coli sepsis ( 5 ).

The presence of urinary reducing substances in the absence of glucosuria (as detected by a routine dipstick test) supports the diagnosis of galactosemia. False-positive and false-negative results are not uncommon. Urinary reducing substances will not be present if the infant is not receiving any dietary galactose (ie, infants already switched to a soy-based formula). The Beutler enzyme spot test is not widely available outside of academic centres but is a more sensitive, rapid test for galactosemia due to GALT deficiency. A recent blood transfusion can cause a false-negative result, while G6PD deficiency results in false-positive results. The confirmatory test (red blood cell quantitative GALT assay) may take some time, but in the interim, a galactose-free diet should be instituted. Finally, red blood cell galactose-1-phosphate concentrations are elevated in all causes of galactosemia regardless of the specific enzyme defect, but the test is not widely available.

In the above case, the discordant glucose meter and serum sample glucose concentrations also provided an important clue to the diagnosis. The need to confirm glucose meter checks with serum samples should be emphasized. Some glucose meters will overestimate the serum glucose in the setting of hypergalactosemia due to the lack of specificity of the enzyme used by the assay ( 6 ). As well, untreated galactosemic infants like the one described above may be hypoglycemic because a major source of their dietary glucose is, in effect, not available.

CLINICAL PEARLS

  • Conjugated hyperbilirubinemia in the newborn is always pathological and requires a systematic approach to diagnosis. Common and treatable conditions should be ruled out promptly.
  • Galactosemia is a relatively rare disorder, but early recognition is key to improving outcome.
  • If galactosemia is suspected, a galactose-free diet should be initiated pending confirmatory test results.
  • Those caring for newborns need to be aware that some commercially available glucose meters will overestimate serum glucose levels in the setting of galactosemia.

ACKNOWLEDGEMENT

Special thanks for the assistance of Dr Joseph Madden, Paediatrician, North Bay General Hospital.

  • Open access
  • Published: 12 June 2024

Risk factors for neonatal hypoxic ischemic encephalopathy and therapeutic hypothermia: a matched case-control study

  • Suoma Roto 1 ,
  • Irmeli Nupponen 2 ,
  • Ilkka Kalliala 1 &
  • Marja Kaijomaa   ORCID: orcid.org/0000-0003-2180-1483 1  

BMC Pregnancy and Childbirth volume  24 , Article number:  421 ( 2024 ) Cite this article

144 Accesses

2 Altmetric

Metrics details

Peripartum asphyxia is one of the main causes of neonatal morbidity and mortality. In moderate and severe cases of asphyxia, a condition called hypoxic-ischemic encephalopathy (HIE) and associated permanent neurological morbidities may follow. Due to the multifactorial etiology of asphyxia, it may be difficult prevent, but in term neonates, therapeutic cooling can be used to prevent or reduce permanent brain damage. The aim of this study was to assess the significance of different antenatal and delivery related risk factors for moderate and severe HIE and the need for therapeutic hypothermia.

We conducted a retrospective matched case-control study in Helsinki University area hospitals during 2013–2017. Newborn singletons with moderate or severe HIE and the need for therapeutic hypothermia were included. They were identified from the hospital database using ICD-codes P91.00, P91.01 and P91.02. For every newborn with the need for therapeutic hypothermia the consecutive term singleton newborn matched by gender, fetal presentation, delivery hospital, and the mode of delivery was selected as a control. Odds ratios (OR) between obstetric and delivery risk factors and the development of HIE were calculated.

Eighty-eight cases with matched controls met the inclusion criteria during the study period. Maternal and infant characteristics among cases and controls were similar, but smoking was more common among cases (aOR 1.46, CI 1.14–1.64, p  = 0.003). The incidence of preeclampsia, diabetes and intrauterine growth restriction in groups was equal. Induction of labour (aOR 3.08, CI 1.18–8.05, p  = 0.02) and obstetric emergencies (aOR 3.51, CI 1.28–9.60, p  = 0.015) were more common in the case group. No difference was detected in the duration of the second stage of labour or the delivery analgesia.

Conclusions

Smoking, induction of labour and any obstetric emergency, especially shoulder dystocia, increase the risk for HIE and need for therapeutic hypothermia. The decisions upon induction of labour need to be carefully weighed, since maternal smoking and obstetric emergencies can hardly be controlled by the clinician.

Peer Review reports

Peripartum asphyxia, generally referred to as birth asphyxia, is one of the main causes of neonatal mortality worldwide [ 1 ]. Approximately three to five newborns per 1000 live births in developed countries are affected by birth asphyxia [ 2 ]. This condition of hypoxia and acidemia can develop gradually during pregnancy and lead to an emergency cesarean section when detected. It can also develop abruptly when complications during labour occur [ 3 ].

The pathophysiology of birth asphyxia and its multifactorial antecedents are well studied and recognized: An increased risk is associated with maternal health problems such as diabetes mellitus, cholestasis of pregnancy, anemia, and hypertension, as well as fetal conditions like intrauterine growth restriction and infections [ 4 , 5 ]. Extensive effort is made to screen and follow-up these mothers and pregnancies with known obstetric risk factors for development of birth asphyxia.

The clinical signs associated with birth asphyxia may be transient and reversible or lead to permanent neurological impairment or death [ 6 ]. A condition called hypoxic-ischemic encephalopathy may follow and, if diagnosed, can further be divided in mild, moderate, and severe [ 7 ]. A quick recovery, normal level of consciousness, mild neurological signs and absence of seizures are typical to a mild HIE, whereas moderate and severe HIE include presence of seizures, multiorgan failure, primitive reflexes and altered level of consciousness and tone [ 7 ]. The diagnosis of severe birth asphyxia is set when the neonate presents with a five-minute Apgar score of 0 to 3 and a pH of 7.0 or less in the umbilical artery blood sample [ 4 ].

In the severe cases of birth asphyxia, HIE predisposes the resuscitated neonate to permanent neurologic morbidities such as cerebral palsy, epilepsy, and developmental delays. The medical intervention to reduce brain damage in term neonates with moderate and severe HIE is therapeutic hypothermia, i.e., cooling of the neonates to around 33 °C for three days [ 8 ].

Despite the high-quality maternal care and the recognition of antenatal risks, birth asphyxia and HIE remain a challenge in perinatal care. Due to the multifactorial nature of fetal distress [ 9 , 10 ], the adverse outcome is not always predictable in risk pregnancies. In addition, many cases of HIE occur unanticipated in low-risk pregnancies.

The aim of this study was to assess the importance of different obstetric risk factors associated with moderate and severe HIE and the need for therapeutic hypothermia in term neonates delivered at the hospitals of Helsinki University Hospital area. We particularly focused on the management protocols of pregnancy and delivery.

This was a retrospective, matched case-control study concerning pregnancies and deliveries in the Helsinki University Hospital area. The same guidelines for follow-up and treatment of pregnancy and delivery are used in all Helsinki University area hospitals. The neonatal intensive care is centralized at the Neonatal Intensive Care Unit (NICU) in Helsinki University Hospital Women’s Clinic. The study period was from January 1, 2013, to December 31, 2017. The treatment of deliveries in the Helsinki University Hospital area was re-organized after a closure of one delivery hospital in late 2017 and the patient record systems was changed in early 2020. Due to the possible bias caused by these factors, years after 2017 were excluded from the study.

The study group consisted of patients who gave birth to asphyxiated singleton neonates with aforementioned symptoms of moderate or severe HIE. Each neonate was born term (one case of 36 6/7 gestation weeks), was admitted to the NICU and offered therapeutic hypothermia for neuroprotection. The indications for hypothermia were admitted from the international guidelines and previous research [ 2 ].

After each delivery with an asphyxiated newborn, the consecutive term singleton, matched by the delivery hospital, fetal gender, presentation (occipital vs. breech), and the mode of delivery (vaginal, assisted vaginal, elective, emergency, and crash cesarean delivery), was selected as a control. An emergency cesarean was defined as a decision-to-delivery-interval of 30 min and a crash cesarean as an immediate delivery after the decision to deliver. Subgroups were formed based on the mode and onset (spontaneous vaginal delivery, induced vaginal delivery, failed induction and cesarean, cesarean) of delivery.

The data for the study was collected from the hospital database (Siemens Obstetrix). All available information concerning fetal and maternal well-being during pregnancy and delivery was collected. This included maternal age and health (pregestational body mass index (BMI), chronic illnesses, medication), gestation at delivery, parity and previous births, and information concerning hospital visits during the ongoing pregnancy. Data on the time of hospital admission and the time of birth in relation to midwife work shifts was also obtained. We considered and tested multiple previously suggested risk factors for HIE or birth asphyxia [ 11 , 12 , 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 22 , 23 , 24 ] and analyzed their possible interactions.

Statistics/analyses

SPSS version 25.0.0 (IBM SPSS Statistics, Armonk, New York) was used to analyze the data. Independent samples t-test and Chi-square test were used for comparing continuous and categorical variables within subgroups, and for testing the independence of variables. Interactions between variables were further assessed with a stratified analysis. ( S10 )

Crude and adjusted odds ratios (OR and aOR respectively) were calculated using logistic regression to estimate associations between different independent variables and the outcome. From the univariate logistic regression, variables with a p  < 0.1 were exported to the multivariable logistic regression analysis. A forward stepwise logistic regression analysis was used to suggest multivariable models. Statistical significance was declared at p  < 0.05 and the CIs were set to 95%. Due to the novel study design and small sample sizes, we saw fit to try out and present two different approaches to the multivariate logistic regression analysis.

We used Benjamini-Hochberg corrected p -values (q-values) in the univariate logistic regression to account for multiple testing [ 25 ]. Missing values of > 5% per category were imputed using the fully conditional specification method and with maximum iterations of 10.

During the study period, 98 355 deliveries took place in Helsinki University area hospitals, the average being 19 671 per year. One hundred and twelve term neonates (including one case of 36 + 6 gestational weeks) presented with moderate to severe HIE and were admitted to the NICU to receive therapeutic hypothermia. The study period incidence of therapeutic hypothermia for signs of moderate to severe HIE in our obstetric population was 1.0/1000.

After excluding twin pregnancies, ninety-seven singleton pregnancies with neonatal HIE and therapeutic hypothermia made up the primary study group. Due to the failure to find matched controls, nine more pregnancies were excluded, leaving us with the final study group of eighty-eight cases.

Altogether, 45.5% of the neonates (40/88) in the study group were born vaginally and 65% (26/40) of them were ventouse-assisted. Two neonates were vaginally born in breech position. Cesarean deliveries constituted 54.5% (48/88) of study group deliveries, including elective, emergency, and crash procedures with the proportion of 2.0% (1/48), 29.2% (14/48), and 68.8% (33/48) respectively. Four emergency cesarean sections were preceded by a failed instrumental delivery. Due to the matching by delivery mode, the proportions were equal in the control group (Fig.  1 : The mode of delivery among cases of HIE and therapeutic hypothermia). The mortality in the study group was 10.2% (9/88). There were no neonatal deaths in the control group.

figure 1

The mode of delivery (%) among cases of hypoxic ischemic encephalopathy and therapeutic hypothermia

Approximately the same proportion of patients in the groups were nulliparous (62.5% vs. 59.1%, p  = 0.576), and no difference was observed in mean maternal age (31.99 vs. 32.96, p  = 0.239) and BMI (24.20 vs. 23.96, p  = 0.467). The mean number of daily cigarettes (1.9 vs. 0.4) was higher ( p  = 0.001) in the study group. No difference was detected in the mean gestational age at delivery (38.89 vs. 40.21 gestational weeks, p  = 0.160) and newborn weight (3458.61 vs. 3472.33 g, p  = 0.898). Post term pregnancy was more common (3.41% vs. 13.64%, p  = 0.024) in the control group (Table  1 ).

There was no difference in the incidence of the most common antenatal complications, such as hypertension or preeclampsia (10.23% vs. 12.50%, p  = 0.797), intrauterine growth restriction (5.68% vs. 7.95%, p  = 0.552), gestational (13.64% vs. 15.91, p  = 0.671) diabetes, type I diabetes (4.55% vs. 3.41%, p  = 0.701) or suspected chorionamniotis (7.95% vs. 6.82%, p  = 0.773). There were no cases of diabetes type II in the study group (0% vs. 2.27%, p  = 0.497) and cholestasis of pregnancy in the control group (3.41% vs. 0%, p  = 0.246) (Table  2 ).

We detected a higher incidence of labour induction in the study group (21.59% vs. 9.09%, p  = 0.025), but no difference was detected in the incidence of cesarean after a failed induction (4.55% vs. 11.36%, p  = 0.106) or the phase II duration of delivery (35.44 min vs. 46.38 min, p  = 0.098). The overall incidence of any obstetric emergency, i.e., shoulder dystocia, placental abruption, or uterine rupture, was higher ( p  = 0.038) in the study group (20.45% vs. 10.23%), driven by a markedly higher incidence of shoulder dystocia (6.82% vs. 0%, p  = 0.029).

There was no difference in the use of epidural (56.82% vs. 68.18%, p  = 0.121), spinal (31.82% vs. 29.55%, p  = 0.744) or oral opioid (20.45% vs. 23.86%, p  = 0.586) anesthesia of deliveries, whereas the use of oxytocin augmentation (27.27% vs. 57,95%, p  < 0.001) and nitrous oxide (38.64% vs. 53.41%, p  = 0.050) was more common in the control group.

Midwife shift change during the active phase of delivery (45.45% vs. 60.23%, p  = 0.051) was somewhat more frequent in the control group and the incidence of delivery during the night shift insignificantly more common in the study group (48.86% vs. 37.50%, p  = 0.070) (Table  2 ).

The univariate analysis showed that nine independent variables were associated ( p  < 0.1) with either the presence or absence of moderate to severe HIE: Smoking, post term pregnancy, induction of delivery, duration of phase II, any obstetric emergency, augmentation of delivery by oxytocin (all stages of labour, including induction), use of nitrous oxide, shift change of midwives during active delivery, and delivery during night shift (10 pm. to 8 am.).

In the multivariate regression model with four to eight variables in the same model, obstetric emergencies, labour induction and smoking significantly increased the odds of HIE (Table  3 , Supporting information Tables S1 - S9 ). We were able to repeat these results in most of the tried models. Induction of labour had a significant association with HIE ( p  = 0.02) in all tried models, but there was no significant association with HIE and the subgroups of induction methods (balloon catheter, vaginal misoprostol, amniotomy followed by oxytocin-infusion), when entered separately to the regression analysis. In fact, in just 33% of cases only one induction method was used.

In the stratified analysis, the association of induction of labour with HIE was even stronger when oxytocin augmentation was used, OR 9.2 (2.71–31.21). Also, the midwife shift change in induced labours resulted in higher OR for HIE (4.5, 1.73–12.20) (Supporting information, Table S10 ). When adjusted with other variables in logistic regression, the significant association of oxytocin use and HIE was still strong, while shift change, duration of the second phase of delivery, and delivery during night shift lost their statistical significance (Table  3 ).

To reveal any common features in different modes of delivery, results were further analyzed in four subgroups: spontaneous and assisted vaginal delivery, and emergency and crash cesarean (Supporting information, Table S11 ). Mothers without preceding active labour or medical intervention, were omitted from the crash cesarean subgroup.

We also made efforts to deeper analyze the cases of shoulder dystocia and induced labours.

There were six cases of shoulder dystocia in the study group, but none in the control group, which made the regression analysis inapplicable for this specific variable. However, the analyses of all obstetric emergencies (placental abruption, uterine rupture, shoulder dystocia) as a surrogate variable showed a statistically significant association with obstetric emergencies and HIE. The increase in odds of HIE with placental abruption and uterine rupture was insignificant or nonexistent. Aforementioned obstetric emergencies altogether presented an OR of 2.57 and aOR of 3.51 ( p  < 0.05) (Table  3 ). Other obstetric emergencies, such as cord prolapse and eclampsia, were not present in our data. The analysis of induced labours showed that even though newborns in the study group were heavier (3790 g vs. 3314 g, p  = 0.030), they were more often born vaginally (84.2% vs. 37.5%, p  = 0.027) (Supporting information, Table S12 ).

When analyzed by the mode of delivery, induction was more common in the study group in vaginal (OR 2.75, 95% 1.13–6.68, p  = 0.016) and assisted (ventouse) vaginal deliveries ( p  = 0.017) (Supporting information, Table S11 ). The midwife shift change was more common in the control groups of the emergency ( p  = 0.008) and crash ( p  = 0.044) caesarean sections and smoking was more common ( p  = 0.039) in the study group of the crash cesarean subgroup. Five of the six study group cases with shoulder dystocia occurred in the ventouse delivery subgroup ( p  = 0.051) (Supporting information Table S11 ).

In this study, maternal smoking, induction of labour and obstetric emergencies appeared to be independent risk factors for HIE. There was a clear dose-dependent association with maternal smoking and HIE. This finding prevailed in the multivariate analysis, although the increase in odds remained quite small. There were more induced labours in the study group and the association with labour induction was most pronounced in the subgroup that received oxytocin, accounting for the use during and after induction. Also shoulder dystocia, a poorly predictable obstetric emergency, increased the risk for HIE. Other previously stated antecedents, e.g., nulliparity, gestational age, maternal weight [ 22 ], prematurity [ 15 ] and chorionamnionitis [ 6 ] appeared mostly not to associate with HIE in this study. Furthermore, post term pregnancy, nitrous oxide, and the use of oxytocin as an independent variable had a seemingly opposite association with HIE.

Smoking is known to be a major risk factor for birth asphyxia and HIE. It is strongly associated with antecedents for asphyxia, i.e., fetal growth restriction [ 26 ] and the risk of placental abruption [ 6 , 27 , 28 ]. Smoking increases oxidative stress and reduces endogenous defenses in the fetus, which may play a role in the pathogenesis [ 29 ]. Even though the harmful effect of smoking is quite indisputable, some bias in the results has to be recognized. The proportion of missing data was substantial, and the imputed data may have skewed the results towards HIE. Also, the frequency, cessation and continuity of smoking was self-reported and susceptible to social desirability bias. It may be, however, safe to assume that the effect of smoking is at least what is presented by the unimputed data (OR 1.21, 95% CI 0.99–1.46, p  = 0.06).

The association between labour induction and HIE requires careful analysis. Significant multicollinearity between induction of labour and other supposed risk factors (obstetric emergency, oxytocin augmentation, shift change, nitrous oxide, and gestational diabetes) was noticed (Supporting information, Table S13 ). The induced labours in the study group ended more frequently in vaginal delivery than in the control group. There were no differences in the indications of labour induction. When these factors are weighed in, the independence of induction of labour as a risk factor for HIE can be considered a complex issue.

The role of induction is, however, worth serious consideration, since these pregnancies may include mothers or fetuses with multiple risk factors. In Finland the rate of induced labours has increased from 17.5% in 2007 to 33.9% in 2021 [ 30 ]. In addition, the proportion of elective inductions without a medical indication are also increasing [ 31 ]. In this study, the risk for HIE was most pronounced among patients with induction of labour together with the use of oxytocin during labour. The oxytocin associated increase in the incidence of encephalopathy was also described in the recent review and meta-analysis by Burgod et al. [ 32 ]. It is also worth noticing that even though newborns in the study group of induced labours were heavier, they were more often born vaginally and the number of ventouse deliveries was twice the proportion in the control group. Compared to zero cases in the study group, in approximately one third of control group cases, a crash cesarean followed a failed ventouse delivery. It can be speculated whether some anchoring bias in decision making is involved and the higher proportions of ventouse and vaginal deliveries in the study group and crash cesareans following ventouse trials in the control group reflect the clinicians’ decisions that are associated with the outcome of the newborn. The number of cases is however too small to draw conclusions.

Shoulder dystocia is an obstetric emergency, that results in prolongation of head-to-body delivery, traction of the brachial plexus, and possible birth trauma [ 33 , 34 ]. The shoulder dystocia incidence reported in studies is approximately 0.7% [ 35 ]. Fetal macrosomia is known to increase the risk of shoulder dystocia more than tenfold [ 35 ] and in these situations, a planned delivery at early term has been demonstrated to reduce the risk of shoulder dystocia [ 34 ]. In this study, six cases of shoulder dystocia were detected in the study group (6.8%) compared to none in the control group. This made the regression analysis inapplicable for this variable. Even though the analysis of all obstetric emergencies (placental abruption, uterine rupture, shoulder dystocia) as a surrogate variable was associated with HIE, the association of HIE with placental abruption and uterine rupture alone was less clear.

As stated, the use of oxytocin in general (irrespective of induction) and nitrous oxide was significantly more common in the control group. However, as shown in the stratified analysis (supplementary information Table S10 ), in the subgroup of induced labours, oxytocin use was more common in the study group. As the need for induction of labour itself may indicate increased risks in the pregnancy, these variables together increase the risk for adverse outcome. In contrast, spontaneous deliveries with oxytocin augmentation were more frequent in the control group. We suggest that the seemingly protective association of oxytocin augmentation in relation to HIE in the regression models could be explained by the asymmetric distribution of these different subgroups. The same can be speculated for the negative association of the administration of nitrous oxide.

The higher incidence of post-term pregnancies in the control group also needs additional attention. It can be speculated that the need for interventions in control group pregnancies was lower and post term was reached more often. It is also of note, that there was significant collinearity between post term pregnancy and oxytocin administration, midwife shift change and delivery during night shift.

In our study population, 54.5% of patients had a cesarean section and the incidences of emergency and crash cesarean were 15.9% and 37.5%. This describes the underlying existence of ante- and intrapartum complications in the study cohort, since the overall incidences of cesarean sections in the Finnish population were 16.7%, 9.2% and 0.9%, respectively [ 30 ]. For example, the rates of pre-eclampsia and pregnancy-induced hypertension in the study were 10.2% and 12.5% compared to our national and worldwide incidences of 5% and 7% [ 36 ].

The purpose of this study was to find HIE risk factors that could be anticipated and avoided in the antenatal care and treatment of delivery. For some patients in the study group, the active labour surveillance, and early obstetric interventions, were never at hand. Our efforts in prevention of HIE should be targeted to patients, that during labour are under constant care and observation.

Compared to previous studies, the selection criteria for this study group were different. Although a similar approach with therapeutic hypothermia as surrogate outcome for severe birth asphyxia (and sequential HIE) has been used before [ 19 ], most case-control studies rest on a study group of neonates diagnosed with neonatal asphyxia, or with signs of birth asphyxia (low Apgar score and/or signs of acidemia in the peripartum blood samples) [ 16 , 18 , 20 , 21 , 24 , 27 , 37 ]. In this study, we chose to use the application of therapeutic hypothermia as the study group inclusion criteria, since it is a clearly defined clinical intervention and in our clinic the indications for use are standardized. The incidence of our inclusion criteria, therapeutic hypothermia (1.1/1000), is slightly higher than the incidence of moderate and severe HIE (0.67/1000) in the study by Liljeström et al. [ 22 ]. Although moderate and severe HIE are the main indications for therapeutic hypothermia, the direct comparison of these incidences should be done with caution. Since the exact severity of HIE may still be uncertain immediately after birth (which may have occurred in another hospital) and the decision concerning this undeniably beneficial treatment has to be made within six hours, the incidence of therapeutic hypothermia treatment may be somewhat higher than the exact incidence of diagnosed moderate and severe HIE.

The study setting could be considered a strength of this study. To the best of our knowledge, this was the first case-control study pairing the groups by the mode of delivery, sex, hospital, and fetal presentation at birth. This could partially explain the differences in our results compared to previous similar studies.

The limitations of this study were its retrospective nature and small sample sizes. It is also likely that the matched case control setting together with a small number of cases failed to show the risks associated with previously described risk factors like hypertension, diabetes, and intrauterine growth restriction. These limitations, as well as coincidence, may also explain the higher incidence of post term pregnancies in the control group. The multicollinearity of some studied risk factors also set limitations when interpreting the data.

There were also some restrictions regarding obtaining data. We didn’t have access to primary health care and antenatal outpatient data, and we relied on the history information of the maternity card and database information upon mothers’ admission to the hospital. Chronic illnesses, obstetric complications and infections were not always structurally recorded. Some information such as substance abuse may be underrepresented but unlikely affects our results.

Demographic risk factors, such as social and marital status, are not collected and had to be excluded. Some previously identified risk factors (urinary tract and viral infections) [ 17 , 23 , 38 , 39 ] had to be excluded because they are treated at the primary health care level.

After controlling for multiple testing, only two of the univariate logistic regression results (maternal smoking and use of oxytocin) remained statistically significant. When studying rare outcomes in limited sample size, one must be careful not to reject the null hypothesis too readily, while minding possibly important findings that fail to reach nominal statistical significance. We considered both these pitfalls and considered clinical applicability as best we could while interpreting these results, but conclusions based on the findings should still be done with caution.

According to our results, induction of labor may be an independent risk factor for HIE, and it should only be used in situations where it evidently improves the outcome of labour. Special vigilance is required from the obstetric team when deciding upon induction and when managing these patients during labour. The increased risk of HIE associated with smoking and obstetric emergencies is unfortunately mostly out of the clinician’s reach.

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Abbreviations

  • Hypoxic-ischemic encephalopathy

Neonatal Intensive Care Unit

Lawn JE, Cousens S, Zupan J. 4 Million neonatal deaths: When? Where? Why? Vol. 365, Lancet.2005;365(9462):891–900.

Jacobs SE, Berg M, Hunt R, Tarnow-Mordi WO, Inder TE, Davis PG. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev. 2013;2013(1):CD003311.

PubMed   PubMed Central   Google Scholar  

Yli BM, Kjellmer I. Pathophysiology of foetal oxygenation and cell damage during labour. Best Pract Res Clin Obstet Gynaecol. 2016;30:9–21.

Article   PubMed   Google Scholar  

Igboanugo S, Chen A, Mielke JG. Maternal risk factors for birth asphyxia in low-resource communities. A systematic review of the literature. J Obstet Gynaecol. 2020;40(8):1039–55.

Rossi AC, Prefumo F. Antepartum and intrapartum risk factors for neonatal hypoxic-ischemic encephalopathy: a systematic review with meta-analysis. Curr Opin Obstet Gynecol. 2019;31(6):410–17.

Sarnat HB, Sarnat MS. Neonatal Encephalopathy following fetal distress: a clinical and electroencephalographic study. Arch Neurol. 1976;33(10):696–705.

Article   CAS   PubMed   Google Scholar  

Morales P, Bustamante D, Espina-Marchant P, Neira-Peña T, Gutiérrez-Hernández MA, Allende-Castro C et al. Pathophysiology of perinatal asphyxia: can we predict and improve individual outcomes? EPMA J;2(2). p. 211–30.

Shankaran S, Laptook AR, Ehrenkranz RA, Tyson JE, McDonald SA, Donovan EF, et al. Whole-body hypothermia for neonates with hypoxic-ischemic encephalopathy. N Engl J Med. 2005;353(15):1574–84.

Westgate JA, Gunn AJ, Gunn TR. Antecedents of neonatal encephalopathy with fetal acidaemia at term. Br J Obstet Gynaecol. 1999;106(8):774–82.

Seikku L, Gissler M, Andersson S, Rahkonen P, Stefanovic V, Tikkanen M, et al. Asphyxia, neurologic morbidity, and perinatal mortality in early- term and postterm birth. Pediatrics. 2016;137(6):e20153334.

Locatelli A, Incerti M, Paterlini G, Doria V, Consonni S, Provero C, et al. Antepartum and intrapartum risk factors for neonatal encephalopathy at term. Am J Perinatol. 2010;27(8):649–54.

Torres-Muñoz J, Rojas C, Mendoza-Urbano D, Marín-Cuero D, Orobio S, Echandía C. Risk factors associated with the development of perinatal asphyxia in neonates at the Hospital Universitario del Valle, Cali, Colombia, 2010–2011. Biomedica. 2017 1;37(0):51–6.

Lundgren C, Brudin L, Wanby AS, Blomberg M. Ante- and intrapartum risk factors for neonatal hypoxic ischemic encephalopathy. J Matern Fetal Neonatal Med. 2018;18(12):1595–601.

Article   Google Scholar  

Itoo BA, Al-Hawsawi ZM, Khan AH. Hypoxic ischemic encephalopathy. Incidence and risk factors in North Western Saudi Arabia. Neurosciences. 2003;8(2):113–9.

PubMed   Google Scholar  

Ayuk Widiani NN, Yuli Kurniati DP, Trisna Windiani IGA. Maternal and infant risk factors on the incidence of neonatal asphyxia in Bali: Case Control Study. Public Health Prev Med Arch. 2016;4(2):120–6.

Hall DR, Smith M, Smith J. Maternal factors contributing to asphyxia neonatorum. J Trop Pediatr. 1996;42(4):192–5.

Badawi N, Kurinczuk JJ, Keogh JM, Alessandri LM, O’Sullivan F, Burton PR, et al. Intrapartum risk factors for newborn encephalopathy: the western Australian case-control study. Br Med J. 1998;317(7172):1549–53.

Article   CAS   Google Scholar  

Blume HK, Loch CM, Li CI. Neonatal encephalopathy and socioeconomic status: Population-based case-control study. Arch Pediatr Adolesc Med. 2007;161(7):663–8.

Nelson DB, Lucke AM, Mcintire DD, Sánchez PJ, Leveno KJ, Chalak LF. Obstetric antecedents to body cooling treatment of the Newborn Infant. Am J Obs Gynecol. 2014;211(2):155–6.

Nayeri F, Shariat M, Dalili H, Bani Adam L, Zareh Mehrjerdi F, Shakeri A. Perinatal risk factors for neonatal asphyxia in Vali-e-Asr hospital, Tehran-Iran. Iran J Reprod Med. 2012;10(2):137–40.

Heinonen S, Saarikoski S. Reproductive risk factors of fetal asphyxia at delivery: a population based analysis. J Clin Epidemiol. 2001;54(4):407–10.

Liljestrom L, Wikstrom AK, Agren J, Jonsson M. Antepartum risk factors for moderate to severe neonatal hypoxic ischemic encephalopathy: a Swedish national cohort study. Acta Obstet Gynecol Scand. 2018;97(5):615–23.

Martinez-Biarge M, Madero R, Gonzlez A, Quero J, Garca-Alix A. Perinatal morbidity and risk of hypoxic-ischemic encephalopathy associated with intrapartum sentinel events. Am J Obstet Gynecol. 2012;206(2):148.e1-148.e7.

Ellis M, Manandhar N, Manandhar DS, De L, Costello AM. Risk factors for neonatal encephalopathy in Kathmandu, Nepal, a developing country: unmatched case-control study. Br Med J. 2000;320(7244):1229–36.

Benjamini Y, Hochberg Y. Controlling the false Discovery rate: a practical and powerful Approach to multiple testing. J R Stat Soc Ser B. 1995;57(1):289–300.

Abraham M, Alramadhan S, Iniguez C, Duijts L, Jaddoe VWV, Den Dekker HT, et al. A systematic review of maternal smoking during pregnancy and fetal measurements with meta-analysis. PLoS ONE Public Libr Sci. 2017;12(2):e0170946.

Tikkanen M, Surcel HM, Bloigu A, Nuutila M, Ylikorkala O, Hiilesmaa V, et al. Self-reported smoking habits and serum cotinine levels in women with placental abruption. Acta Obstet Gynecol Scand. 2010;89(12):1538–44.

Downes KL, Shenassa ED, Grantz KL. Neonatal outcomes Associated with placental abruption. Am J Epidemiol. 2017;186(12):1319–28.

Article   PubMed   PubMed Central   Google Scholar  

Gitto E, Reiter RJ, Karbownik M, Tan D-x, Gitto P, Berberi S, et al. Causes of oxidative stress in the pre- and Perinatal Period. Neonatology. 2002;81(3):146–57.

Perinataalitilasto. – synnyttäjät, synnytykset ja vastasyntyneet - THL [Internet]. [cited 2020 Aug 6]. https://thl.fi/fi/tilastot-ja-data/tilastot-aiheittain/seksuaali-ja-lisaantymisterveys/synnyttajat-synnytykset-ja-vastasyntyneet/perinataalitilasto-synnyttajat-synnytykset-ja-vastasyntyneet .

Kruit H, Nuutila M, Rahkonen L. Lääkärilehti - Synnytyksen käynnistäminen, Kun raskaus on täysiaikainen. Lääkärilehti. 2016;71(25–32/2016):1845–51.

Google Scholar  

Burgod C, Pant S, Morales MM, Montaldo P, Ivain P, Elangovan R, et al. Effect of intra-partum oxytocin on neonatal encephalopathy: a systematic review and meta-analysis. BMC Pregnancy Childbirth. 2021;21(1):1–7.

Collins KA, Popek E. Birth Injury: Birth Asphyxia and Birth Trauma. Academic Forensic Pathology. Volume 8. SAGE Publications Inc.; 2018. pp. 788–864.

Farrar D, Simmonds M, Bryant M, Sheldon TA, Tuffnel D, Golder S, et al. Treatments for gestational diabetes: a systematic review and meta-analysis. BMJ Open. 2017;7(6):e015557.

Hartiadystokia. - Duodecim Oppiportti [Internet]. [cited 2020 Aug 18]. https://www-oppiportti-fi.libproxy.helsinki.fi/op/njs15507/do?p_haku=hartiadystokia#q=hartiadystokia .

Rana S, Lemoine E, Granger J, Karumanchi SA, Preeclampsia. Pathophysiology, challenges, and perspectives. Circ Res. 2019;124(7):1094–112.

Milsom I, Ladfors L, Thiringer K, Niklasson A, Odeback A, Thornberg E. Influence of maternal, obstetric and fetal risk factors on the prevalence of birth asphyxia at term in a Swedish urban population. Acta Obstet Gynecol Scand. 2002;81(10):909–17.

Hayes BC, McGarvey C, Mulvany S, Kennedy J, Geary MP, Mattherws TG et al. A case-control study of hypoxic-ischemic encephalopathy in newborn infants at > 36 weeks gestation. Am J Obstet Gynecol. 2013;209(1):29.e1-29.e19.

Kapaya H, Williams R, Elton G, Anumba D. Can obstetric risk factors predict fetal acidaemia at Birth? A retrospective case-control study. J Pregnancy. 2018;2018:2195965.

Download references

Acknowledgements

We gratefully acknowledge the assistance of Paula Bergman, biostatistician at Biostatistics consulting, University of Helsinki, Finland, for her biostatistical advice.

Helsinki University State Research Funding.

Open Access funding provided by University of Helsinki (including Helsinki University Central Hospital).

Author information

Authors and affiliations.

Department of obstetrics and gynecology, Helsinki University Women’s Hospital, Haartmaninkatu 2, Helsinki, 00029, Finland

Suoma Roto, Ilkka Kalliala & Marja Kaijomaa

Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland

Irmeli Nupponen

You can also search for this author in PubMed   Google Scholar

Contributions

SR contributed to the literature search, figures, study design, data collection, data analysis, data interpretation and writing. IN contributed to the study design, data collection data interpretation and writing. IK contributed to the data interpretation and writing. MK contributed to the study design, data collection, data analysis, data interpretation, figures, and writing.

Corresponding author

Correspondence to Marja Kaijomaa .

Ethics declarations

Ethical approval.

The study was approved by the Helsinki University Hospital Institutional Review Board. (March 3, 2018, 1/2018).

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ . The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Cite this article.

Roto, S., Nupponen, I., Kalliala, I. et al. Risk factors for neonatal hypoxic ischemic encephalopathy and therapeutic hypothermia: a matched case-control study. BMC Pregnancy Childbirth 24 , 421 (2024). https://doi.org/10.1186/s12884-024-06596-8

Download citation

Received : 31 May 2023

Accepted : 20 May 2024

Published : 12 June 2024

DOI : https://doi.org/10.1186/s12884-024-06596-8

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Birth asphyxia
  • Induction of labour
  • Oxytocin treatment
  • Shoulder dystocia
  • Therapeutic hypothermia

BMC Pregnancy and Childbirth

ISSN: 1471-2393

case study neonatal hyperbilirubinemia

IMAGES

  1. Hyperbilirubinemia Case Study

    case study neonatal hyperbilirubinemia

  2. Neonatal Hyperbilirubinemia

    case study neonatal hyperbilirubinemia

  3. PPT

    case study neonatal hyperbilirubinemia

  4. Unconjugated Neonatal Hyperbilirubinemia: Complications

    case study neonatal hyperbilirubinemia

  5. 2 MIN MEDICINE|Neonatal hyperbilirubinemia

    case study neonatal hyperbilirubinemia

  6. (PDF) Risk Factors and Outcome of Neonatal Hyperbilirubinemia: A Case

    case study neonatal hyperbilirubinemia

VIDEO

  1. NEONATOLOGY lecture 7 JAUNDICE NEONATORUM unconjugated hyperbilirubinemia in detail

  2. Rare case of Neonatal progeroid syndrome

  3. Neonatal Hyperbilirubinemia

  4. Panel discussion Neonatal Hyperbilirubinemia

  5. part 1 ||Neonatal jaundice|| icterus neonatorum|| neonatal hyperbilirubinemia || types ||Hindi

  6. NEONATAL HYPERBILIRUBINEMIA BY DR. PRAJAWAL POUDEL

COMMENTS

  1. Case 1: Severe Jaundice in a 2-day-old Term Neonate

    A 2-day-old, 2.68-kg term male neonate is brought to the emergency department with lethargy, poor feeding, and significant generalized jaundice. He was born via spontaneous vaginal delivery at home to a gravida 4, para 3 Amish woman under the supervision of a midwife, at an estimated gestational age of 39 weeks after an uncomplicated pregnancy with scant prenatal care. Jaundice was noticed 7 ...

  2. Hyperbilirubinemia in Neonates: Types, Causes, Clinical Examinations

    Neonatal hyperbilirubinemia is a common clinical problem encountered during the neonatal period, ... In case of breastfed newborns, mild jaundice may take 10-14 days after birth or may reoccur during the breast feeding period ... Erythroblastosis fetalis, VIII: studies of serum bilirubin in relation to kernicterus. N Engl J Med, 247: 668-71.

  3. Clinical Practice Guideline Revision: Management of Hyperbilirubinemia

    More than 80% of newborn infants will have some degree of jaundice. 1,2 Careful monitoring of all newborn infants and the application of appropriate treatments are essential, because high bilirubin concentrations can cause acute bilirubin encephalopathy and kernicterus. 3 Kernicterus is a permanent disabling neurologic condition characterized by some or all of the following: choreoathetoid ...

  4. Neonatal Jaundice

    Neonatal jaundice is a clinical manifestation of elevated total serum bilirubin (TSB), termed neonatal hyperbilirubinemia, which results from bilirubin that is deposited into an infant's skin. The characteristic features of neonatal jaundice include yellowish skin, sclerae, and mucous membranes. Jaundice derives from the French word jaune, meaning yellow. Neonatal jaundice is the most ...

  5. Technical Report: Diagnosis and Management of Hyperbilirubinemia in the

    PubMed.STUDY SELECTION. English language randomized controlled trials and observational studies. Excluded: case reports or series, nonsystematic reviews, and investigations focused on <35-weeks' gestation infants.DATA EXTRACTION. ... that evaluated the association of neonatal jaundice and neonatal phototherapy with childhood allergic diseases.

  6. Evaluation of Newborn Hyperbilirubinemia and Jaundice in Primary Care

    This case report serves as a comprehensive exploration of a newborn presenting with increased jaundice in the primary care setting. It emphasizes the critical role of clinicians in promptly addressing abnormal bilirubin results in newborn patients and effectively monitoring and managing hyperbilirubinemia after hospital discharge. By providing clinical considerations for practice and practical ...

  7. Assessment, management, and incidence of neonatal jaundice in ...

    Kuzniewicz, M. W. et al. Risk factors for severe hyperbilirubinemia among infants with borderline bilirubin levels: A nested case-control study. J. Pediatr. 153 , 234-240.

  8. Case 34-2019: A 16-Year-Old Boy with Jaundice

    Dr. Akash Gupta (Pediatrics): A 16-year-old boy was admitted to this hospital in autumn because of jaundice and abnormal results on liver-function tests. The patient had been well until 4 days ...

  9. Clinical and genetic risk factors associated with neonatal severe

    Objective: We aimed to investigate the clinical and genetic risk factors associated with neonatal severe unconjugated hyperbilirubinemia. Methods: This was a retrospective, 1:1 matched, case-control study. We included 614 neonates diagnosed with severe unconjugated hyperbilirubinemia (serum total bilirubin level ≥425 μmol/L or serum total bilirubin concentration that met exchange ...

  10. Management of Neonatal Hyperbilirubinemia: Shedding Light on the

    Neonatal hyperbilirubinemia and bilirubin neurotoxicity in hospitalized neonates: analysis of the US Database. Pediatr Res. 2022; 91(7):1662-1668. 10.1038/s41390-021-01692-3 PMID: 34429513 > Crossref Medline Google Scholar; 19. Meek JY, Noble L. Technical report: breast-feeding and the use of human milk. Pediatrics.

  11. Neonatal Hyperbilirubinemia: Evaluation and Treatment

    The AAP neonatal hyperbilirubinemia guideline provides hour-specific thresholds for initiation of phototherapy based on gestational age and the presence or absence of neurotoxicity risk factors. 4 ...

  12. Nursing Care of Neonatal Jaundice in Hyperbilirubinemia Babies: a Case

    This study aims to describe nursing care for neonatal jaundice in hyperbilirubinemia infants. Methods: This study uses a case study approach design through the nursing process with a sample of a ...

  13. Risk factors for severe hyperbilirubinemia in neonates

    The incidence of severe neonatal hyperbilirubinemia is higher in Asians than in whites. A case-control study was designed to investigate the effects of eight known risk factors [breast feeding, ABO incompatibility, premature birth, infection, cephalohematoma, asphyxia, glucose-6-phosphate dehydrogenase (G6PD) deficiency, and variant UDP-glucuronosyltransferase 1A1 (UGT1A1) gene] and a ...

  14. Diagnostic methods for neonatal hyperbilirubinemia: benefits

    Invasive bilirubin measurements remain the gold standard for the diagnosis and treatment of infants with severe neonatal hyperbilirubinemia. The present paper describes different methods currently ...

  15. Maternal and neonatal factors associated with neonatal jaundice in

    This study aimed to identify maternal and neonatal factors associated with neonatal jaundice among hospitalised neonates in north Jordan. Methods This was a case control study involving 312 mothers and their neonates (106 cases, 206 controls), recruited from two governmental hospitals in north Jordan.

  16. (PDF) Neonatal jaundice causes and management

    Keywords: Neonatal jaundice, Neonatal hyperbilirubinemia, Prematurity, Kernicter us . 1 Taif University, ... opening the door for further case studies in medical applications, especially in ...

  17. Recent advances in the management of neonatal jaundice

    Introduction. Neonatal hyperbilirubinemia is the most common clinical condition in the newborn requiring evaluation and management and remains a frequent reason for hospital readmission during the first week of postnatal life. 1, Citation 2 The high prevalence of neonatal hyperbilirubinemia reflects developmental red blood cell, hepatic, and gastrointestinal immaturities that result in an ...

  18. Clinical features and genetic variations of severe neonatal

    Case 1: The neonate was diagnosed with neonatal hyperbilirubinemia and CHPS. Case 2: He was diagnosed as G6PD deficiency, neonatal hyperbilirubinemia, ... We propose that the two genetic variations may aggravate the jaundice levels in this case. A previous study noted that spherical-shaped red cells were rarely observed in neonates, ...

  19. PDF Stony Brook University Hospital

    Stony Brook University Hospital

  20. Neonatal jaundice incidence, risk factors and outcomes in 54 referral

    The case fatality rate for jaundice was 2.6% (22/845). ... 20 A scoping review of neonatal jaundice studies in Nigeria (1960-2014) found that one in five neonatal admissions were due to significant neonatal hyperbilirubinemia. 20 The current study found around 15% of special care baby unit admissions were due to jaundice, ...

  21. Case Study: Neonatal Jaundice

    Case Background. Neonatal jaundice in a disorder that affects nearly 50% of all newborns to at least a small degree. The yellow coloration of the skin and sclera of the eyes is due to the accumulation of bilirubin in adipose tissue and its adherence to collagen fibers.

  22. 4 Hyperbilirubinemia (Neonatal Jaundice) Nursing Care Plans

    A study reported that 70.8% of late preterm neonates and 29.1% of term neonates has at least one neonatal morbidity like neonatal jaundice, hypoglycemia, respiratory morbidities, and sepsis. They observed jaundice in 55.1% of late preterm neonates who required phototherapy, and hypoglycemia was found in 8.8% of late preterm neonates (Salman et ...

  23. Preventive effect of prenatal maternal oral probiotic supplementation

    Introduction Neonatal jaundice is a common and life-threatening health problem in neonates due to overaccumulation of circulating unconjugated bilirubin. Gut flora has a potential influence on bilirubin metabolism. The infant gut microbiome is commonly copied from the maternal gut. During pregnancy, due to changes in dietary habits, hormones and body weight, maternal gut dysbiosis is common ...

  24. Risk factors for neonatal hyperbilirubinemia: a systematic review and

    A previous study showed that the cases of neonatal hyperbilirubinemia caused by ABO blood group incompatibility accounted for 13.3 . In two Chinese studies, maternal-fetal blood group incompatibility was considered an independent risk factor for neonatal jaundice (18,19). There are some limitations to our research.

  25. Case study of neonatal jaundice

    The document describes a medical case report for a 3-day-old female infant admitted to the NICU for neonatal jaundice. She presented with yellowish discoloration of the skin and eyes. Her vital signs were normal except for occasional hypothermia. Her physical exam found jaundice, poor nutrition, and abnormal neurological responses.

  26. The effect of phototherapy treatment on serum melatonin levels in term

    This study was planned as a single-center, prospective, observational, case-control study. Term infants (gestation week ≥37 weeks) who received at least 6 h of phototherapy due to jaundice constitute the phototherapy group, while the term infants ... neonatal hyperbilirubinemia was first reported in 1958 by Cremer et al. [25]. Despite its ...

  27. Jaundice: Causes, Symptoms, and Treatment Options

    Jaundice, also known as icterus, is the yellowish discoloration of the skin and eyes caused by the abnormal buildup of an orangish waste product called bilirubin. Bilirubin is produced by the normal breakdown of red blood cells (RBCs) and processed by the liver, where it is cleared from the body in bile.If too much bilirubin is produced or the liver cannot clear it, jaundice can develop.

  28. Case 2: Newborn with jaundice and "hyperglycemia"

    A nine-day-old boy was admitted to the paediatric ward of a regional hospital after a maternal-newborn clinic appointment, where he was found to be jaundiced with a total serum bilirubin of 400 μmol/L. He had been followed by the public health nurse in the community for poor weight gain in his first week of life.

  29. Risk factors for neonatal hypoxic ischemic encephalopathy and

    Although a similar approach with therapeutic hypothermia as surrogate outcome for severe birth asphyxia (and sequential HIE) has been used before , most case-control studies rest on a study group of neonates diagnosed with neonatal asphyxia, or with signs of birth asphyxia (low Apgar score and/or signs of acidemia in the peripartum blood ...