1
|
Cornet MC, Numis AL, Monsell SE, Chan NH, Gonzalez FF, Comstock BA, Juul SE, Wusthoff CJ, Wu YW, Glass HC. Assessing Early Severity of Hypoxic-Ischemic Encephalopathy: The Role of Electroencephalogram Background in Addition to Sarnat Exam. J Pediatr 2025; 277:114411. [PMID: 39557386 DOI: 10.1016/j.jpeds.2024.114411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/07/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
OBJECTIVE To assess the relationship between the Sarnat exam, early electroencephalogram (EEG) background, and death or neurodevelopmental impairment (NDI) at age 2 years among neonates with moderate to severe hypoxic-ischemic encephalopathy treated with therapeutic hypothermia. STUDY DESIGN Neonates enrolled in the High-dose Erythropoietin for Asphyxia and Encephalopathy trial with EEG (n = 463) or amplitude-integrated electroencephalogram (n = 15) reports available on the first day after birth were included in this cohort study. A Sarnat exam was performed between 1 and 6 hours after birth, and neonates were classified into 3 groups of increasing severity based on the number of severe features (none, 1-2, or 3+). EEG background continuity was extracted from reports and categorized as normal, excessively discontinuous, or severely abnormal. The primary outcome was severe NDI or death at age 2. RESULTS Among 478 neonates with hypoxic-ischemic encephalopathy, EEG background continuity was normal in 186 (39%), excessively discontinuous in 171 (36%), and severely abnormal in 121 (25%). For each additional severe feature on the Sarnat exam, the risk of abnormal EEG background increased by 16% (relative risk 1.16 [95% CI 1.09-1.23]). Both the Sarnat exam and EEG background severity were associated with an increased risk of severe NDI or death. After adjusting for Sarnat exam severity, severe EEG background remained associated with severe NDI and death (relative risk 5.7 [95% CI 3.7-8.9]). CONCLUSIONS The early EEG background provides additional information beyond the Sarnat exam and could be an additional early marker when assessing the severity of HIE.
Collapse
Affiliation(s)
- Marie-Coralie Cornet
- Department of Pediatrics, University of California San Francisco, San Francisco, CA.
| | - Adam L Numis
- Department of Neurology and the Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Sarah E Monsell
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Natalie H Chan
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Fernando F Gonzalez
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Bryan A Comstock
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Sandra E Juul
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | | | - Yvonne W Wu
- Department of Pediatrics, University of California San Francisco, San Francisco, CA; Department of Neurology and the Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Hannah C Glass
- Department of Pediatrics, University of California San Francisco, San Francisco, CA; Department of Neurology and the Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA
| |
Collapse
|
2
|
Abend NS, Wusthoff CJ, Jensen FE, Inder TE, Volpe JJ. Neonatal Seizures. VOLPE'S NEUROLOGY OF THE NEWBORN 2025:381-448.e17. [DOI: 10.1016/b978-0-443-10513-5.00015-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Agarwal N, Benedetti GM. Neuromonitoring in the ICU: noninvasive and invasive modalities for critically ill children and neonates. Curr Opin Pediatr 2024; 36:630-643. [PMID: 39297699 DOI: 10.1097/mop.0000000000001399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
PURPOSE OF REVIEW Critically ill children are at risk of neurologic dysfunction and acquiring primary and secondary brain injury. Close monitoring of cerebral function is crucial to prevent, detect, and treat these complications. RECENT FINDINGS A variety of neuromonitoring modalities are currently used in pediatric and neonatal ICUs. These include noninvasive modalities, such as electroencephalography, transcranial Doppler, and near-infrared spectroscopy, as well as invasive methods including intracranial pressure monitoring, brain tissue oxygen measurement, and cerebral microdialysis. Each modality offers unique insights into neurologic function, cerebral circulation, or metabolism to support individualized neurologic care based on a patient's own physiology. Utilization of these modalities in ICUs results in reduced neurologic injury and mortality and improved neurodevelopmental outcomes. SUMMARY Monitoring of neurologic function can significantly improve care of critically ill children. Additional research is needed to establish normative values in pediatric patients and to standardize the use of these modalities.
Collapse
Affiliation(s)
- Neha Agarwal
- Division of Pediatric Neurology, Department of Pediatrics, University of Michigan, C.S. Mott Children's Hospital, Ann Arbor, Michigan, USA
| | | |
Collapse
|
4
|
Mathieson SR, Nanyunja C, Sadoo S, Nakalembe S, Duckworth E, Muryasingura S, Niombi N, Proietti J, Busingye M, Nakimuli A, Livingstone V, Webb EL, Mambule I, Boylan GB, Tann CJ. EEG background activity, seizure burden and early childhood outcomes in neonatal encephalopathy in Uganda: a prospective feasibility cohort study. EClinicalMedicine 2024; 78:102937. [PMID: 39640940 PMCID: PMC11617306 DOI: 10.1016/j.eclinm.2024.102937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Background Intrapartum-related neonatal encephalopathy (NE) is a leading cause of childhood mortality and morbidity. Continuous electroencephalography (EEG) is gold standard for neonatal brain monitoring; however, low-income country data is lacking. We examined EEG in a Ugandan cohort with NE to describe feasibility, background activity, seizure prevalence and burden, and associations with clinical presentation and outcome. Methods Neonates with NE were recruited from a single hospital referral centre in Kampala, Uganda (Oct 2019-Oct 2020) and underwent EEG monitoring. Feasibility was assessed as to whether EEG monitoring of diagnostic quality could be achieved from days 1-5. Evolution of clinical presentation was assessed by Sarnat classification and daily Thompson score was performed. EEG background severity was graded at 12, 24, 48 and 72 h after birth, and at time of Thompson score. Seizures were annotated remotely by experts and assessed for frequency, duration, burden, and status epilepticus. Early childhood outcome was assessed at follow up, and adverse outcome defined as death or neurodevelopmental impairment (NDI) at 18-24 months of age. Findings In this prospective feasibility cohort study, diagnostic quality EEGs were recorded for 50 of 51 recruited neonates (median duration 71.4 h, IQR 52.4-72.2), indicating feasibility. Of 39 participants followed to 18-24 months, 13 died and 7 had NDI. Daily Thompson score and EEG background grade were strongly correlated across all timepoints (days 1-5). Thompson score of ≥7 was most predictive of moderate-severe EEG background abnormality (AUC 0.83). Prognostic accuracy of moderate-severe EEG background grade to predict NDI was high (AUC 0.74). Electrographic seizures were seen in 52% (26); median seizure burden was high at 264 min (IQR 27.8-523.7, range 1.3-1374.1); half (13) had status epilepticus. Interpretation EEG monitoring was feasible as a research tool in this sub-Saharan Africa setting. EEG background activity correlated strongly with scored neurological assessment and predicted adverse early childhood outcome. Seizure prevalence and burden, including status epilepticus, were high in this uncooled cohort with important potential longer-term implications for survivors. Funding Bill & Melinda Gates Foundation grant number OPP1210890; Wellcome Trust Innovator award (209325/Z/17/Z).
Collapse
Affiliation(s)
- Sean R. Mathieson
- INFANT Research Centre and Department of Paediatrics & Child Health, University College Cork, Ireland
| | - Carol Nanyunja
- MRC/UVRI & LSHTM Uganda Research Unit, Entebbe
- London School of Hygiene & Tropical Medicine, Keppel Street, London
| | - Samantha Sadoo
- London School of Hygiene & Tropical Medicine, Keppel Street, London
- University College London Hospitals NHS Trust, Euston Road, London
| | | | | | | | | | - Jacopo Proietti
- INFANT Research Centre and Department of Paediatrics & Child Health, University College Cork, Ireland
| | | | - Annettee Nakimuli
- Kawempe National Referral Hospital, Kampala, Uganda
- Makerere University, Kampala, Uganda
| | - Vicki Livingstone
- INFANT Research Centre and Department of Paediatrics & Child Health, University College Cork, Ireland
| | - Emily L. Webb
- London School of Hygiene & Tropical Medicine, Keppel Street, London
| | | | - Geraldine B. Boylan
- INFANT Research Centre and Department of Paediatrics & Child Health, University College Cork, Ireland
| | - Cally J. Tann
- MRC/UVRI & LSHTM Uganda Research Unit, Entebbe
- London School of Hygiene & Tropical Medicine, Keppel Street, London
- University College London Hospitals NHS Trust, Euston Road, London
| |
Collapse
|
5
|
Edoigiawerie S, Henry J, Issa N, David H. A Systematic Review of EEG and MRI Features for Predicting Long-Term Neurological Outcomes in Cooled Neonates With Hypoxic-Ischemic Encephalopathy (HIE). Cureus 2024; 16:e71431. [PMID: 39539899 PMCID: PMC11558949 DOI: 10.7759/cureus.71431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) represents a significant global disease burden, but more importantly, it leaves a lasting impact of disability on individual children and their families. HIE outcome prognostication is important for guiding clinical interventions and counseling families. The objective of this study was to systematically review early electroencephalogram (EEG) and magnetic resonance imaging (MRI) features associated with long-term neurological outcomes in infants after perinatal HIE. Articles were extracted from PubMed, CINAHL, and Scopus. Twenty studies were included that assessed EEG and/or MRI patterns in neonates who underwent therapeutic hypothermia and were followed to determine long-term outcomes. Articles that did not meet the inclusion criteria were excluded. Covidence review manager (Melbourne, Australia: Covidence) was used to extract, evaluate, and synthesize review results. Of the articles included, eight focused on EEG features, eight on MRI features, and four on assessments using both EEG and MRI. Abnormal EEG background and burst suppression severity were associated with poor outcomes. Higher MRI injury scores in the basal ganglia and thalamus were also correlated with poor outcomes. Finally, studies also revealed restricted diffusion and greater lesion size in the subcortical gray matter correlated with poor outcomes. We also identified limitations in the included studies which primarily involved sample size, potential for MRI pseudonormalization, and the potential tradeoff between retention of infants able to receive long-term follow-up and attrition of those lost to follow-up. We conclude that EEG background patterns, MRI scoring, subcortical lesion burden, and MRI diffusivity are sensitive metrics for predicting outcomes. Both early EEG and MRI features may serve as high-fidelity biomarkers for secondary energy failure and for counseling families of neonates at high risk for devastating neurologic outcomes. Additionally, there is a paucity of information on the impact of HIE on brain areas outside of the standard clinical basal-ganglia and watershed patterns, especially in locations like the corpus callosum. Finally, MRI pseudonormalization may underestimate the extent of injury in these studies.
Collapse
Affiliation(s)
| | - Julia Henry
- Pediatric Neurology, AdventHealth Medical Group, Orlando, USA
| | - Naoum Issa
- Neurological Surgery, University of Chicago Medical Center, Chicago, USA
| | - Henry David
- Pediatric Neurology, University of Chicago Medical Center, Chicago, USA
| |
Collapse
|
6
|
Ryan MAJ, Malhotra A. Electrographic monitoring for seizure detection in the neonatal unit: current status and future direction. Pediatr Res 2024; 96:896-904. [PMID: 38684885 PMCID: PMC11502487 DOI: 10.1038/s41390-024-03207-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 03/20/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024]
Abstract
Neonatal neurocritical intensive care is dedicated to safeguarding the newborn brain by prioritising clinical practices that promote early identification, diagnosis and treatment of brain injuries. The most common newborn neurological emergency is neonatal seizures, which may also be the initial clinical indication of neurological disease. A high seizure burden in the newborn period independently contributes to increased mortality and morbidity. The majority of seizures in newborns are subclinical (without clinical presentation), and hence identification may be difficult. Neuromonitoring techniques most frequently used to monitor brain wave activity include conventional electroencephalography (cEEG) or amplitude-integrated EEG (aEEG). cEEG with video is the gold standard for diagnosing and treating seizures. Many neonatal units do not have access to cEEG, and frequently those that do, have little access to real-time interpretation of monitoring. IMPACT: EEG monitoring is of no benefit to an infant without expert interpretation. Whilst EEG is a reliable cot-side tool and of diagnostic and prognostic use, both conventional EEG and amplitude-integrated EEG have strengths and limitations, including sensitivity to seizure activity and ease of interpretation. Automated seizure detection requires a sensitive and specific algorithm that can interpret EEG in real-time and identify seizures, including their intensity and duration.
Collapse
Affiliation(s)
- Mary Anne J Ryan
- INFANT Research Centre, University College Cork, Cork, Ireland.
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| | - Atul Malhotra
- Monash Newborn, Monash Children's Hospital, Melbourne, Australia
- Department of Paediatrics, Monash University, Melbourne, Australia
| |
Collapse
|
7
|
Proietti J, O'Toole JM, Murray DM, Boylan GB. Advances in Electroencephalographic Biomarkers of Neonatal Hypoxic Ischemic Encephalopathy. Clin Perinatol 2024; 51:649-663. [PMID: 39095102 DOI: 10.1016/j.clp.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Electroencephalography (EEG) is a key objective biomarker of newborn brain function, delivering critical, cotside insights to aid the management of encephalopathy. Access to continuous EEG is limited, forcing reliance on subjective clinical assessments. In hypoxia ischaemia, the primary cause of encephalopathy, alterations in EEG patterns correlate with. injury severity and evolution. As HIE evolves, causing secondary neuronal death, EEG can track injury progression, informing neuroprotective strategies, seizure management and prognosis. Despite its value, challenges with interpretation and lack of on site expertise has limited its broader adoption. Technological advances, particularly in digital EEG and machine learning, are enhancing real-time analysis. This will allow EEG to expand its role in HIE diagnosis, management and outcome prediction.
Collapse
Affiliation(s)
- Jacopo Proietti
- Department of Engineering for Innovation Medicine, University of Verona, Strada le Grazie, Verona 37134, Italy; INFANT Research Centre, University College Cork, Cork, Ireland
| | - John M O'Toole
- INFANT Research Centre, University College Cork, Cork, Ireland; Cergenx Ltd., Dublin, Ireland
| | - Deirdre M Murray
- INFANT Research Centre, University College Cork, Cork, Ireland; Department of Paediatrics & Child Health, University College Cork, Paediatric Academic Unit, Cork University Hospital, Wilton, Cork, T12 DC4A, Ireland
| | - Geraldine B Boylan
- INFANT Research Centre, University College Cork, Cork, Ireland; Department of Paediatrics & Child Health, University College Cork, Paediatric Academic Unit, Cork University Hospital, Wilton, Cork, T12 DC4A, Ireland.
| |
Collapse
|
8
|
Zhou KQ, Dhillon SK, Bennet L, Davidson JO, Gunn AJ. How do we reach the goal of personalized medicine for neuroprotection in neonatal hypoxic-ischemic encephalopathy? Semin Perinatol 2024; 48:151930. [PMID: 38910063 DOI: 10.1016/j.semperi.2024.151930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Therapeutic hypothermia is now standard of care for neonates with hypoxic-ischemic encephalopathy (HIE) in high income countries (HIC). Conversely, compelling trial evidence suggests that hypothermia is ineffective, and may be deleterious, in low- and middle-income countries (LMIC), likely reflecting the lower proportion of infants who had sentinel events at birth, suggesting that injury had advanced to a stage when hypothermia is no longer effective. Although hypothermia significantly reduced the risk of death and disability in HICs, many infants survived with disability and in principle may benefit from targeted add-on neuroprotective or neurorestorative therapies. The present review will assess biomarkers that could be used to personalize treatment for babies with HIE - to determine first whether an individual infant is likely to respond to hypothermia, and second, whether additional treatments may be beneficial.
Collapse
Affiliation(s)
- Kelly Q Zhou
- Dept of Physiology, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Simerdeep K Dhillon
- Dept of Physiology, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Laura Bennet
- Dept of Physiology, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Joanne O Davidson
- Dept of Physiology, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Alistair J Gunn
- Dept of Physiology, The University of Auckland, Private Bag 92019, Auckland, New Zealand.
| |
Collapse
|
9
|
Roychaudhuri S, Hannon K, Sunwoo J, Garvey AA, El-Dib M. Quantitative EEG and prediction of outcome in neonatal encephalopathy: a review. Pediatr Res 2024; 96:73-80. [PMID: 38503980 DOI: 10.1038/s41390-024-03138-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/18/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024]
Abstract
Electroencephalogram (EEG) is an important biomarker for neonatal encephalopathy (NE) and has significant predictive value for brain injury and neurodevelopmental outcomes. Quantitative analysis of EEG involves the representation of complex EEG data in an objective, reproducible and scalable manner. Quantitative EEG (qEEG) can be derived from both a limited channel EEG (as available during amplitude integrated EEG) and multi-channel conventional EEG. It has the potential to enable bedside clinicians to monitor and evaluate details of cortical function without the necessity of continuous expert input. This is particularly useful in NE, a dynamic and evolving condition. In these infants, continuous, detailed evaluation of cortical function at the bedside is a valuable aide to management especially in the current era of therapeutic hypothermia and possible upcoming neuroprotective therapies. This review discusses the role of qEEG in newborns with NE and its use in informing monitoring and therapy, along with its ability to predict imaging changes and short and long-term neurodevelopmental outcomes. IMPACT: Quantitative representation of EEG data brings the evaluation of continuous brain function, from the neurophysiology lab to the NICU bedside and has a potential role as a biomarker for neonatal encephalopathy. Clinical and research applications of quantitative EEG in the newborn are rapidly evolving and a wider understanding of its utility is valuable. This overview summarizes the role of quantitative EEG at different timepoints, its relevance to management and its predictive value for short- and long-term outcomes in neonatal encephalopathy.
Collapse
Affiliation(s)
- Sriya Roychaudhuri
- Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Katie Hannon
- Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - John Sunwoo
- Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Aisling A Garvey
- Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital, Boston, MA, USA
- INFANT Research Centre, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Mohamed El-Dib
- Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Hua T, Nguyen TT, Nguyen TT. Progression of Amplitude-Integrated Electroencephalography and Neurological Outcomes in Neonates With Hypoxic-Ischemic Encephalopathy: A Single-Institution Cohort Study in Vietnam. Cureus 2024; 16:e62317. [PMID: 38882227 PMCID: PMC11177235 DOI: 10.7759/cureus.62317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 06/18/2024] Open
Abstract
Background The characteristics of amplitude-integrated electroencephalography (aEEG) are associated with neurological outcomes in neonates with hypoxic-ischemic encephalopathy (HIE). We perform a longitudinal analysis of continuous monitoring of aEEG during therapeutic hypothermia and explore the association between aEEG interpretation and clinical neurological outcomes. Method We conducted a prospective cohort study on HIE neonates undergoing hypothermia with aEEG monitoring. Results A total of 37 HIE infants underwent hypothermia with improved aEEG background activity in 28 (75.7%) neonates, of which 18 (48.6%) neonates had background activity returned to a continuous pattern, and the median recovery time was 26.5 hours. Sleep-wake cycle (SWC) appeared in 14 (37.8%) cases, with a median onset time of 34.5 hours. Seizure activity on aEEG was present in 26 (70.3%) infants. Factors associated with poor outcomes at discharge included low voltage or flat trace background activity, a lack of improvement in background activity after hypothermia, and the absence of SWC. Neonates who took longer than 62 hours to return to continuous background activity (time to normal trace) or did not have SWC before the end of hypothermia were more likely to have unfavorable outcomes at discharge. Conclusions Longitudinal analysis of aEEG during hypothermia should be implemented in neonatal care units. The progression of these features on aEEG may predict neurological outcomes for HIE neonates.
Collapse
Affiliation(s)
- Thu Hua
- Department of Pediatrics, Children's Hospital 2, Ho Chi Minh, VNM
| | - Thien T Nguyen
- Department of Pediatrics and Neonatology, Children's Hospital 2, Ho Chi Minh, VNM
| | - Tinh T Nguyen
- Department of Pediatrics, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh, VNM
- Department of Neonatology, Children's Hospital 2, Ho Chi Minh, VNM
- Department of Neonatology, Ho Chi Minh University Medical Center, Ho Chi Minh, VNM
| |
Collapse
|
11
|
Hung SC, Tu YF, Hunter SE, Guimaraes C. MRI predictors of long-term outcomes of neonatal hypoxic ischaemic encephalopathy: a primer for radiologists. Br J Radiol 2024; 97:1067-1077. [PMID: 38407350 PMCID: PMC11654721 DOI: 10.1093/bjr/tqae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/12/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024] Open
Abstract
This review aims to serve as a foundational resource for general radiologists, enhancing their understanding of the role of Magnetic Resonance Imaging (MRI) in early prognostication for newborns diagnosed with hypoxic ischaemic encephalopathy (HIE). The article explores the application of MRI as a predictive instrument for determining long-term outcomes in newborns affected by HIE. With HIE constituting a leading cause of neonatal mortality and severe long-term neurodevelopmental impairments, early identification of prognostic indicators is crucial for timely intervention and optimal clinical management. We examine current literature and recent advancements to provide an in-depth overview of MRI predictors, encompassing brain injury patterns, injury scoring systems, spectroscopy, and diffusion imaging. The potential of these MRI biomarkers in predicting long-term neurodevelopmental outcomes and the probability of epilepsy is also discussed.
Collapse
Affiliation(s)
- Sheng-Che Hung
- Department of Radiology, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, United States
| | - Yi-Fang Tu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Senyene E Hunter
- Department of Neurology, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599-7025, United States
| | - Carolina Guimaraes
- Department of Radiology, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|
12
|
Lavrentev SN, Petrova AS, Serova OF, Vishnyakova P, Kondratev MV, Gryzunova AS, Zakharova NI, Zubkov VV, Silachev DN. Ultrasound Diagnosis and Near-Infrared Spectroscopy in the Study of Encephalopathy in Neonates Born under Asphyxia: Narrative Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:591. [PMID: 38790586 PMCID: PMC11119551 DOI: 10.3390/children11050591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024]
Abstract
Brain injury resulting from adverse events during pregnancy and delivery is the leading cause of neonatal morbidity and disability. Surviving neonates often suffer long-term motor, sensory, and cognitive impairments. Birth asphyxia is among the most common causes of neonatal encephalopathy. The integration of ultrasound, including Doppler ultrasound, and near-infrared spectroscopy (NIRS) offers a promising approach to understanding the pathology and diagnosis of encephalopathy in this special patient population. Ultrasound diagnosis can be very helpful for the assessment of structural abnormalities associated with neonatal encephalopathy such as alterations in brain structures (intraventricular hemorrhage, infarcts, hydrocephalus, white matter injury) and evaluation of morphologic changes. Doppler sonography is the most valuable method as it provides information about blood flow patterns and outcome prediction. NIRS provides valuable insight into the functional aspects of brain activity by measuring tissue oxygenation and blood flow. The combination of ultrasonography and NIRS may produce complementary information on structural and functional aspects of the brain. This review summarizes the current state of research, discusses advantages and limitations, and explores future directions to improve applicability and efficacy.
Collapse
Affiliation(s)
- Simeon N. Lavrentev
- The State Budgetary Institution, Moscow Regional Perinatal Center, 143912 Balashikha, Russia; (S.N.L.); (A.S.P.); (O.F.S.); (M.V.K.); (A.S.G.)
- Research Clinical Institute of Childhood of the Moscow Region, 115093 Moscow, Russia; (N.I.Z.); (V.V.Z.)
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia;
| | - Anastasia S. Petrova
- The State Budgetary Institution, Moscow Regional Perinatal Center, 143912 Balashikha, Russia; (S.N.L.); (A.S.P.); (O.F.S.); (M.V.K.); (A.S.G.)
- Research Clinical Institute of Childhood of the Moscow Region, 115093 Moscow, Russia; (N.I.Z.); (V.V.Z.)
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia;
| | - Olga F. Serova
- The State Budgetary Institution, Moscow Regional Perinatal Center, 143912 Balashikha, Russia; (S.N.L.); (A.S.P.); (O.F.S.); (M.V.K.); (A.S.G.)
| | - Polina Vishnyakova
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia;
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Maxim V. Kondratev
- The State Budgetary Institution, Moscow Regional Perinatal Center, 143912 Balashikha, Russia; (S.N.L.); (A.S.P.); (O.F.S.); (M.V.K.); (A.S.G.)
- Research Clinical Institute of Childhood of the Moscow Region, 115093 Moscow, Russia; (N.I.Z.); (V.V.Z.)
| | - Anastasia S. Gryzunova
- The State Budgetary Institution, Moscow Regional Perinatal Center, 143912 Balashikha, Russia; (S.N.L.); (A.S.P.); (O.F.S.); (M.V.K.); (A.S.G.)
- Research Clinical Institute of Childhood of the Moscow Region, 115093 Moscow, Russia; (N.I.Z.); (V.V.Z.)
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia;
| | - Nina I. Zakharova
- Research Clinical Institute of Childhood of the Moscow Region, 115093 Moscow, Russia; (N.I.Z.); (V.V.Z.)
| | - Victor V. Zubkov
- Research Clinical Institute of Childhood of the Moscow Region, 115093 Moscow, Russia; (N.I.Z.); (V.V.Z.)
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia;
| | - Denis N. Silachev
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia;
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
13
|
Keene JC, Loe ME, Fulton T, Keene M, Mathur A, Morrissey MJ, Tomko SR, Vesoulis ZA, Zempel JM, Ching S, Guerriero RM. Macroperiodic Oscillations: A Potential Novel Biomarker of Outcome in Neonatal Encephalopathy. J Clin Neurophysiol 2024; 41:344-350. [PMID: 37052470 PMCID: PMC10567988 DOI: 10.1097/wnp.0000000000001011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
PURPOSE Neonatal encephalopathy (NE) is a common cause of neurodevelopmental morbidity. Tools to accurately predict outcomes after therapeutic hypothermia remain limited. We evaluated a novel EEG biomarker, macroperiodic oscillations (MOs), to predict neurodevelopmental outcomes. METHODS We conducted a secondary analysis of a randomized controlled trial of neonates with moderate-to-severe NE who underwent standardized clinical examination, magnetic resonance (MR) scoring, video EEG, and neurodevelopmental assessment with Bayley III evaluation at 18 to 24 months. A non-NE cohort of neonates was also assessed for the presence of MOs. The relationship between clinical examination, MR score, MOs, and neurodevelopmental assessment was analyzed. RESULTS The study included 37 neonates with 24 of whom survived and underwent neurodevelopmental assessment (70%). The strength of MOs correlated with severity of clinical encephalopathy. MO strength and spread significantly correlated with Bayley III cognitive percentile ( P = 0.017 and 0.046). MO strength outperformed MR score in predicting a combined adverse outcome of death or disability ( P = 0.019, sensitivity 100%, specificity 77% vs. P = 0.079, sensitivity 100%, specificity 59%). CONCLUSIONS MOs are an EEG-derived, quantitative biomarker of neurodevelopmental outcome that outperformed a comprehensive validated MRI injury score and a detailed systematic discharge examination in this small cohort. Future work is needed to validate MOs in a larger cohort and elucidate the underlying pathophysiology of MOs.
Collapse
Affiliation(s)
- Jennifer C Keene
- Division of Pediatric and Developmental Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, U.S.A
| | - Maren E Loe
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, Missouri, U.S.A
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, Missouri, U.S.A
| | - Talie Fulton
- Washington University in St. Louis, St. Louis, Missouri, U.S.A
| | - Maire Keene
- Division of Pediatric and Developmental Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, U.S.A
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, Missouri, U.S.A
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, Missouri, U.S.A
- Washington University in St. Louis, St. Louis, Missouri, U.S.A
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, U.S.A. ; and
- Division of Newborn Medicine, Department of Pediatrics. Washington University in St. Louis, St. Louis, Missouri, U.S.A
| | - Amit Mathur
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, U.S.A. ; and
| | - Michael J Morrissey
- Division of Pediatric and Developmental Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, U.S.A
| | - Stuart R Tomko
- Division of Pediatric and Developmental Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, U.S.A
| | - Zachary A Vesoulis
- Division of Newborn Medicine, Department of Pediatrics. Washington University in St. Louis, St. Louis, Missouri, U.S.A
| | - John M Zempel
- Division of Pediatric and Developmental Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, U.S.A
| | - ShiNung Ching
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, Missouri, U.S.A
| | - Réjean M Guerriero
- Division of Pediatric and Developmental Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, U.S.A
| |
Collapse
|
14
|
Yuliati A, Zayek M, Maertens P. The Impact of Phenobarbital on the Ability of Electroencephalogram to Predict Adverse Outcome in Asphyxiated Neonates during Therapeutic Hypothermia. Am J Perinatol 2024; 41:e1681-e1688. [PMID: 37186086 DOI: 10.1055/s-0043-1768487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
OBJECTIVE Classification of electroencephalogram (EEG) background has been established to predict outcome in neonates with hypoxic ischemic encephalopathy (HIE). However, the impact of phenobarbital therapy on the predictability of EEG background has not been studied. Our objective is to determine if EEG background after treatment with phenobarbital during therapeutic hypothermia (TH) remains a good predictor for brain injury in neonates with HIE. STUDY DESIGN This is a single-center, retrospective study of consecutive neonates with HIE who underwent TH and EEG monitoring from October 2017 to March 2021. Per institutional protocol, all infants received a dose of prophylactic phenobarbital and bumetanide therapy at the onset of TH for sedative and neuroprotective measures. The initial 3 hours of EEG background activity was classified based on national guidelines. Infants were separated into two groups based on EEG background scores: group 1 (normal-mild, n = 30) and group 2 (moderate-severe, n = 36). Brain magnetic resonance imaging (MRI) results were scored based on the National Institute of Child Health and Human Development (NICHD) criteria. Adverse outcomes were defined as death before MRI or NICHD brain injury score > 1A. RESULTS Infants in group 2 had lower Apgar scores at 5 minutes of age, severe acidemia, moderate to severe encephalopathy score, and earlier initiation of EEG monitoring than infants in group 1. Moderate to severe EEG background score was associated with presence of brain injury on MRI or death (p = 0.003), and this association remained significant even after adjustment for independent risk factors (odds ratio = 56.24 [95% confidence interval = 1.841-1718], p = 0.021). CONCLUSION Phenobarbital therapy does not affect the ability of EEG to predict adverse outcome in infants with perinatal asphyxia during TH. KEY POINTS · EEG has a clinical utility for predicting outcome in neonates with hypoxia-ischemia.. · Phenobarbital therapy is commonly used in neonates, and may impact EEG background findings.. · In spite phenobarbital therapy, moderate to severe EEG background abnormalities in infants with perinatal asphyxia during TH remain an excellent predictor for poor outcome..
Collapse
Affiliation(s)
- Asri Yuliati
- Division of Pediatric Neurology, Department of Pediatrics, University of South Alabama, Mobile, Alabama
| | - Michael Zayek
- Department of Pediatrics, Division of Neonatology, University of South Alabama, Mobile, Alabama
| | - Paul Maertens
- Division of Pediatric Neurology, Department of Pediatrics, University of South Alabama, Mobile, Alabama
| |
Collapse
|
15
|
Sakpichaisakul K, El-Dib M, Munster C, Supapannachart KJ, Yang E, Walsh BH, Volpe JJ, Inder TE. Amplitude-Integrated Electroencephalography Evolution and Magnetic Resonance Imaging Injury in Mild and Moderate to Severe Neonatal Encephalopathy. Am J Perinatol 2024; 41:e2463-e2473. [PMID: 37369240 DOI: 10.1055/a-2118-2953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
OBJECTIVE This study aimed to describe the evolution of amplitude-integrated electroencephalography (aEEG) in neonatal encephalopathy (NE) during therapeutic hypothermia (TH) and evaluate the association between aEEG parameters and magnetic resonance imaging (MRI) injury. STUDY DESIGN aEEG data of infants who underwent TH were reviewed for background, sleep wake cycling (SWC), and seizures. Conventional electroencephalography (cEEG) background was assessed from the reports. Discordance of background on aEEG and cEEG was defined if there was a difference in the severity of the background. MRI injury (total score ≥ 5) was assessed by using the Weeke scoring system. RESULTS A total of 46 infants were included; 23 (50%) with mild NE and 23 (50%) with moderate to severe NE. Comparing mild NE with moderate to severe NE, the initial aEEG background differed with more mild being continuous (70 vs. 52%), with fewer being discontinuous (0 vs. 22%) and flat tracing (0 vs. 4%), whereas burst suppression (4 vs. 4%) and low voltage (26 vs. 18%) did not differ. There was a notably common discordance between the background assessment on cEEG with aEEG in 82% with continuous and 40% low voltage aEEG background. MRI abnormalities were identified in four infants with mild NE and seven infants with moderate to severe NE. MRI injury was associated with aEEG seizures in infants with moderate to severe NE. CONCLUSION aEEG seizures are useful to predict MRI injury in moderate to severe NE infants. There is a large discrepancy between aEEG, cEEG, and MRI in neonates treated by TH. KEY POINTS · MRI injury was identified in 29% of moderate NE infants and in 50% of severe NE infants.. · aEEG seizures were associated with MRI injury in the moderate to severe NE infants.. · MRI injury was identified in 16% infants with mild NE.. · Mild NE infants with normal aEEG were unlikely to have MRI injury.. · There was a large discrepancy between aEEG, cEEG, and MRI in infants treated by TH..
Collapse
Affiliation(s)
- Kullasate Sakpichaisakul
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Pediatrics, Division of Neurology, Queen Sirikit National Institute of Child Health, Ministry of Public Health, College of Medicine, Rangsit University, Bangkok, Thailand
| | - Mohamed El-Dib
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chelsea Munster
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Krittin J Supapannachart
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Edward Yang
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Brain H Walsh
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Joseph J Volpe
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Langeslag JF, Onland W, Groenendaal F, de Vries LS, van Kaam AH, de Haan TR. Association Between Seizures and Neurodevelopmental Outcome at Two and Five Years in Asphyxiated Newborns With Therapeutic Hypothermia. Pediatr Neurol 2024; 153:152-158. [PMID: 38387280 DOI: 10.1016/j.pediatrneurol.2024.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024]
Abstract
OBJECTIVE To investigate the association between the presence and severity of seizures in asphyxiated newborns and their neurodevelopmental outcome at ages two and five years. METHODS Retrospective data analysis from a prospectively collected multicenter cohort of 186 term-born asphyxiated newborns undergoing therapeutic hypothermia (TH) in 11 centers in the Netherlands and Belgium. Seizures were diagnosed by amplitude-integrated electroencephalography (EEG) and raw EEG signal reading up to 48 hours after rewarming. Neurodevelopmental outcome was assessed by standardized testing at age two and five years. Primary outcome was death or long-term neurodevelopmental impairment (NDI) including cerebral palsy. Associations were calculated using univariate and multivariate logistic regression analyses adjusting for Thompson score and a validated brain magnetic resonance imaging (MRI) score. RESULTS Seventy infants (38%) had seizures during TH or rewarming, and 44 (63%) of these needed two or more antiseizure medications (ASMs). Overall mortality was 21%. Follow-up data from 147 survivors were available for 137 infants (93%) at two and for 94 of 116 infants (81%) at five years. NDI was present in 26% at two and five years. Univariate analyses showed a significant association between seizures and death or NDI, but this was no longer significant after adjusting for Thompson and MRI score in the multivariate analysis; this was also true for severe seizures (need for two or more ASMs) or seizures starting during rewarming. CONCLUSION The presence or severity of seizures in newborns undergoing TH for hypoxic-ischemic encephalopathy was not independently associated with death or NDI up to age five years after adjusting for several confounders.
Collapse
Affiliation(s)
- Juliette F Langeslag
- Department of Neonatology, Amsterdam UMC Location University of Amsterdam, Emma Children's Hospital, Amsterdam, the Netherlands; Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
| | - Wes Onland
- Department of Neonatology, Amsterdam UMC Location University of Amsterdam, Emma Children's Hospital, Amsterdam, the Netherlands; Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, and Brain Center, Utrecht, the Netherlands
| | - Linda S de Vries
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, and Brain Center, Utrecht, the Netherlands
| | - Anton H van Kaam
- Department of Neonatology, Amsterdam UMC Location University of Amsterdam, Emma Children's Hospital, Amsterdam, the Netherlands; Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands
| | - Timo R de Haan
- Department of Neonatology, Amsterdam UMC Location University of Amsterdam, Emma Children's Hospital, Amsterdam, the Netherlands; Amsterdam Reproduction & Development Research Institute, Amsterdam, the Netherlands.
| |
Collapse
|
17
|
Glass HC, Numis AL, Comstock BA, Gonzalez FF, Mietzsch U, Bonifacio SL, Massey S, Thomas C, Natarajan N, Mayock DE, Sokol GM, Van Meurs KP, Ahmad KA, Maitre N, Heagerty PJ, Juul SE, Wu YW, Wusthoff CJ. Association of EEG Background and Neurodevelopmental Outcome in Neonates With Hypoxic-Ischemic Encephalopathy Receiving Hypothermia. Neurology 2023; 101:e2223-e2233. [PMID: 37816642 PMCID: PMC10727206 DOI: 10.1212/wnl.0000000000207744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/20/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Predicting neurodevelopmental outcome for neonates with hypoxic-ischemic encephalopathy (HIE) is important for clinical decision-making, care planning, and parent communication. We examined the relationship between EEG background and neurodevelopmental outcome among children enrolled in a trial of erythropoietin or placebo for neonates with HIE treated with therapeutic hypothermia. METHODS Participants had EEG recorded throughout hypothermia. EEG background was classified as normal, discontinuous, or severely abnormal (defined as burst suppression, low voltage suppressed, or status epilepticus) at 5 1-hour epochs: onset of recording, 24, 36, 48, and 72 hours after birth. The predominant background pattern during the entire continuous video EEG monitoring recording was calculated using the arithmetic mean of the 5 EEG background ratings (normal = 0; discontinuous = 1; severely abnormal = 2) as follows: "predominantly normal" (mean = 0), "normal/discontinuous" (0 < mean<1), "predominantly discontinuous" (mean = 1), "discontinuous/severely abnormal" (1 < mean<2), or "predominantly severely abnormal" (mean = 2). Primary outcome was death or neurodevelopmental impairment (NDI) defined as cerebral palsy, Gross Motor Function Classification Score ≥1, or cognitive score <90 on Bayley Scales of Infant Toddler Development, third edition at age 2 years. Neurodevelopment was also categorized into a 5-level ordinal measure: no, mild, moderate, severe NDI, or death for secondary analysis. We used generalized linear regression models with robust standard errors to assess the relative risk of death or NDI by EEG background in both unadjusted and adjusted analyses controlling for the effects of treatment group, sex, HIE severity, and study recruitment site. RESULTS Among 142 neonates, the predominant background EEG pattern was predominantly normal in 35 (25%), normal/discontinuous in 68 (48%), predominantly discontinuous in 11 (7.7%), discontinuous/severely abnormal in 16 (11%), and predominantly severely abnormal in 12 (8.5%). Increasing severity of background across monitoring epochs was associated with increasingly worse clinical outcomes. Children with severe EEG background abnormality at any time point (n = 36, 25%) were significantly more likely to die or have severe NDI at 2 years (adjusted relative risk: 7.95, 95% CI 3.49-18.12). DISCUSSION EEG background is strongly associated with NDI at age 2 years. These results can be used to assist health care providers to plan follow-up care and counsel families for decision-making related to goals of care.
Collapse
Affiliation(s)
- Hannah C Glass
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA.
| | - Adam L Numis
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Bryan A Comstock
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Fernando F Gonzalez
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Ulrike Mietzsch
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Sonia Lomeli Bonifacio
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Shavonne Massey
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Cameron Thomas
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Niranjana Natarajan
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Dennis E Mayock
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Gregory M Sokol
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Krisa P Van Meurs
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Kaashif A Ahmad
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Nathalie Maitre
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Patrick J Heagerty
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Sandra E Juul
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Yvonne W Wu
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| | - Courtney J Wusthoff
- Departments of Neurology and Weill Institute for Neuroscience (H.C.G., A.L.N., Y.W.W.); Pediatrics (H.C.G., A.L.N., Y.W.W.), UCSF Benioff Children's Hospital; Epidemiology & Biostatistics (H.C.G.), University of California San Francisco, CA; Department Biostatistics (B.A.C., P.J.H.), University of Washington, Seattle; Department of Pediatrics (U.M., S.E.J.), Division of Neonatology, University of Washington School of Medicine, Seattle Children's Hospital; Department of Pediatrics (K.P.V.M., S.L.B.), Division of Neonatal and Developmental Medicine, Stanford University, Palo Alto, CA; Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine (S.L.M.), University of Pennsylvania, Philadelphia; Department of Pediatrics (C.T.), University of Cincinnati and Division of Neurology, Cincinnati Children's Hospital Medical Center, OH; Department of Neurology (N.N.), University of Washington School of Medicine, Seattle; Department of Pediatrics (G.S.), Indiana University School of Medicine, Indianapolis, IN; Pediatrix Neonatology of San Antonio (K.A.A.), TX; Department of Pediatrics, and Emory + Children's Pediatric Institute (N.M.), Emory University, Atlanta, GA; Department of Neurology (C.J.W.), Stanford University, Palo Alto, CA
| |
Collapse
|
18
|
Yadav B, Madaan P, Meena J, Kumar J, Sahu JK. Treatment of electrographic seizures versus clinical seizures in neonates: A systematic review and meta-analysis. Epilepsy Res 2023; 198:107244. [PMID: 39491917 DOI: 10.1016/j.eplepsyres.2023.107244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 10/12/2023] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Electrographic seizures in neonates are commonly associated with poor neurodevelopmental outcomes. However, there is conflict in evidence whether control of electrographic seizures translate into improved neurodevelopmental outcome. We aimed to evaluate whether treating all electrographic seizures compared with treating clinical seizures leads to a better neurodevelopmental outcome at 18-24 months in neonates. METHODS We searched four electronic databases (Medline, Cochrane Library, Embase, and Web of Science) for randomized controlled trials (RCTs) published until October 31, 2022. We included RCTs comparing the treatment of electrographic seizures with clinical seizures in neonates with or at risk of seizures. The Risk of Bias version 2 tool was used to assess the risk of bias. The random-effects meta-analysis was performed using RevMan 5.4 software. RESULTS Of the 2872 unique records identified by the database search, five trials involving 474 infants were eligible for the review. There was no significant difference in the incidence of neurodevelopmental disability at 18-24 months. Infants in the electrographic seizure group had lower scores in the cognitive domain (2 trials, 186 participants, MD: -5.4; 95% CI:-10.3 to -0.4, I2-0%). There were no significant differences in scores in the motor and language domains, mortality, seizure burden, brain injury scores, antiseizure medication use, and post-neonatal epilepsy. CONCLUSIONS Having limitations of small number of trials and heterogeneity, the systematic review does not identify evidence demonstrating improved neurodevelopmental outcomes or improved survival in neonates treated for electrographic seizures versus clinical seizures.
Collapse
Affiliation(s)
- Bharti Yadav
- Department of Neonatology, Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Priyanka Madaan
- Department of Pediatric Neurology, Amrita Institute of Medical Sciences, Faridabad, India
| | - Jitendra Meena
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Jogender Kumar
- Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Jitendra Kumar Sahu
- Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
19
|
Kang OH, Jahn P, Eichhorn JG, Dresbach T, Müller A, Sabir H. Correlation of Different MRI Scoring Systems with Long-Term Cognitive Outcome in Cooled Asphyxiated Newborns. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1295. [PMID: 37628294 PMCID: PMC10453158 DOI: 10.3390/children10081295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023]
Abstract
(1) Background: Cerebral MRI plays a significant role in assessing the extent of brain injury in neonates with neonatal encephalopathy after perinatal asphyxia. Over the last decades, several MRI scoring systems were developed to enhance the predictive accuracy of MRI. The aim of this study was to validate the correlation of four established MRI scoring systems with cognitive long-term outcomes in cooled asphyxiated newborns. (2) Methods: Forty neonates with neonatal encephalopathy treated with therapeutic hypothermia were included in this retrospective study. The MRI scans from the second week of life were scored using four existing MRI scoring systems (Barkovich, NICHD, Rutherford, and Weeke). The patients' outcome was assessed with the Bayley Scales of Infant Development (BSID-III) at the age of 2 years. To evaluate the correlation between the MRI scoring system with the cognitive scores of BSID-III, the correlation coefficient was calculated for each scoring system. (3) Results: All four MRI scoring systems showed a significant correlation with the cognitive scores of BSID-III. The strongest correlation was found between the Weeke Score (r2 = 0.43), followed by the Rutherford score (r2 = 0.39), the NICHD score (r2 = 0.22), and the Barkovich score (r2 = 0.17). (4) Conclusion: Our study confirms previously published results in an independent cohort and indicates that the Weeke and Rutherford scores have the strongest correlation with the cognitive score of BSID-III in cooled asphyxiated newborns.
Collapse
Affiliation(s)
- Ok-Hap Kang
- Children’s Hospital, Klinikum Leverkusen, 51375 Leverkusen, Germany; (O.-H.K.); (P.J.); (J.G.E.)
| | - Peter Jahn
- Children’s Hospital, Klinikum Leverkusen, 51375 Leverkusen, Germany; (O.-H.K.); (P.J.); (J.G.E.)
| | - Joachim G. Eichhorn
- Children’s Hospital, Klinikum Leverkusen, 51375 Leverkusen, Germany; (O.-H.K.); (P.J.); (J.G.E.)
| | - Till Dresbach
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital University of Bonn, 53127 Bonn, Germany; (T.D.); (A.M.)
| | - Andreas Müller
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital University of Bonn, 53127 Bonn, Germany; (T.D.); (A.M.)
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital University of Bonn, 53127 Bonn, Germany; (T.D.); (A.M.)
| |
Collapse
|
20
|
Juul SE, Voldal E, Comstock BA, Massaro AN, Bammler TK, Mayock DE, Heagerty PJ, Wu YW, Numis AL. Association of High-Dose Erythropoietin With Circulating Biomarkers and Neurodevelopmental Outcomes Among Neonates With Hypoxic Ischemic Encephalopathy: A Secondary Analysis of the HEAL Randomized Clinical Trial. JAMA Netw Open 2023; 6:e2322131. [PMID: 37418263 PMCID: PMC10329214 DOI: 10.1001/jamanetworkopen.2023.22131] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/18/2023] [Indexed: 07/08/2023] Open
Abstract
Importance The ability to predict neurodevelopmental impairment (NDI) for infants diagnosed with hypoxic ischemic encephalopathy (HIE) is important for parental guidance and clinical treatment as well as for stratification of patients for future neurotherapeutic studies. Objectives To examine the effect of erythropoietin on plasma inflammatory mediators in infants with moderate or severe HIE and to develop a panel of circulating biomarkers that improves the projection of 2-year NDI over and above the clinical data available at the time of birth. Design, Setting, and Participants This study is a preplanned secondary analysis of prospectively collected data from infants enrolled in the High-Dose Erythropoietin for Asphyxia and Encephalopathy (HEAL) Trial, which tested the efficacy of erythropoietin as an adjunctive neuroprotective therapy to therapeutic hypothermia. The study was conducted at 17 academic sites comprising 23 neonatal intensive care units in the United States between January 25, 2017, and October 9, 2019, with follow-up through October 2022. Overall, 500 infants born at 36 weeks' gestation or later with moderate or severe HIE were included. Intervention Erythropoietin treatment 1000 U/kg/dose on days 1, 2, 3, 4 and 7. Main Outcomes and Measures Plasma erythropoietin was measured in 444 infants (89%) within 24 hours after birth. A subset of 180 infants who had plasma samples available at baseline (day 0/1), day 2, and day 4 after birth and either died or had 2-year Bayley Scales of Infant Development III assessments completed were included in the biomarker analysis. Results The 180 infants included in this substudy had a mean (SD) gestational age of 39.1 (1.5) weeks, and 83 (46%) were female. Infants who received erythropoietin had increased concentrations of erythropoietin at day 2 and day 4 compared with baseline. Erythropoietin treatment did not alter concentrations of other measured biomarkers (eg, difference in interleukin [IL] 6 between groups on day 4: -1.3 pg/mL; 95% CI, -4.8 to 2.0 pg/mL). After adjusting for multiple comparisons, we identified 6 plasma biomarkers (C5a, interleukin [IL] 6, and neuron-specific enolase at baseline; IL-8, tau, and ubiquitin carboxy-terminal hydrolase-L1 at day 4) that significantly improved estimations of death or NDI at 2 years compared with clinical data alone. However, the improvement was only modest, increasing the AUC from 0.73 (95% CI, 0.70-0.75) to 0.79 (95% CI, 0.77-0.81; P = .01), corresponding to a 16% (95% CI, 5%-44%) increase in correct classification of participant risk of death or NDI at 2 years. Conclusions and Relevance In this study, erythropoietin treatment did not reduce biomarkers of neuroinflammation or brain injury in infants with HIE. Circulating biomarkers modestly improved estimation of 2-year outcomes. Trial Registration ClinicalTrials.gov Identifier: NCT02811263.
Collapse
|
21
|
Alharbi HM, Pinchefsky EF, Tran MA, Salazar Cerda CI, Parokaran Varghese J, Kamino D, Widjaja E, Mamak E, Ly L, Nevalainen P, Hahn CD, Tam EWY. Seizure Burden and Neurologic Outcomes After Neonatal Encephalopathy. Neurology 2023; 100:e1976-e1984. [PMID: 36990719 PMCID: PMC10186227 DOI: 10.1212/wnl.0000000000207202] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/03/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Seizures are common during neonatal encephalopathy (NE), but the contribution of seizure burden (SB) to outcomes remains controversial. This study aims to examine the relationship between electrographic SB and neurologic outcomes after NE. METHODS This prospective cohort study recruited newborns ≥36 weeks postmenstrual age around 6 hours of life between August 2014 and November 2019 from a neonatal intensive care unit (NICU). Participants underwent continuous electroencephalography for at least 48 hours, brain MRI within 3-5 days of life, and structured follow-up at 18 months. Electrographic seizures were identified by board-certified neurophysiologists and quantified as total SB and maximum hourly SB. A medication exposure score was calculated based on all antiseizure medications given during NICU admission. Brain MRI injury severity was classified based on basal ganglia and watershed scores. Developmental outcomes were measured using the Bayley Scales of Infant Development, Third Edition. Multivariable regression analyses were performed, adjusting for significant potential confounders. RESULTS Of 108 enrolled infants, 98 had continuous EEG (cEEG) and MRI data collected, of which 5 were lost to follow-up, and 6 died before age 18 months. All infants with moderate-severe encephalopathy completed therapeutic hypothermia. cEEG-confirmed neonatal seizures occurred in 21 (24%) newborns, with a total SB mean of 12.5 ± 36.4 minutes and a maximum hourly SB mean of 4 ± 10 min/h. After adjusting for MRI brain injury severity and medication exposure, total SB was significantly associated with lower cognitive (-0.21, 95% CI -0.33 to -0.08, p = 0.002) and language (-0.25, 95% CI -0.39 to -0.11, p = 0.001) scores at 18 months. Total SB of 60 minutes was associated with 15-point decline in language scores and 70 minutes for cognitive scores. However, SB was not significantly associated with epilepsy, neuromotor score, or cerebral palsy (p > 0.1). DISCUSSION Higher SB during NE was independently associated with worse cognitive and language scores at 18 months, even after adjusting for exposure to antiseizure medications and severity of brain injury. These observations support the hypothesis that neonatal seizures occurring during NE independently contribute to long-term outcomes.
Collapse
Affiliation(s)
- Huda M Alharbi
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Elana F Pinchefsky
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - My-An Tran
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Carlos Ivan Salazar Cerda
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Jessy Parokaran Varghese
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Daphne Kamino
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Elysa Widjaja
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Eva Mamak
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Linh Ly
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland.
| | - Päivi Nevalainen
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Cecil D Hahn
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland
| | - Emily W Y Tam
- From the Department of Pediatrics (H.M.A.), King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Department of Pediatrics (E.F.P.), Centre Hospitalier Universitaire Sainte-Justine and the University of Montreal, Quebec; Neurosciences and Mental Health Program (M.-A.T., J.P.V., E.W., C.D.H., E.W.Y.T.), Hospital for Sick Children Research Institute; Department of Paediatrics (C.I.S.C., D.K., E.W., L.L., C.D.H., E.W.Y.T.), Department of Radiology (E.W.), and Department of Psychology (E.M.), The Hospital for Sick Children and the University of Toronto, Ontario, Canada; and Epilepsia Helsinki (P.N.), Department of Clinical Neurophysiology, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Finland.
| |
Collapse
|
22
|
Fang XY, Tian YL, Chen SY, Shi Q, Zheng D, Wang YJ, Mao J. [A novel method for electroencephalography background analysis in neonates with hypoxic-ischemic encephalopathy]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:128-134. [PMID: 36854687 DOI: 10.7499/j.issn.1008-8830.2208102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
OBJECTIVES To explore a new method for electroencephalography (EEG) background analysis in neonates with hypoxic-ischemic encephalopathy (HIE) and its relationship with clinical grading and head magnetic resonance imaging (MRI) grading. METHODS A retrospective analysis was performed for the video electroencephalography (vEEG) and amplitude-integrated electroencephalography (aEEG) monitoring data within 24 hours after birth of neonates diagnosed with HIE from January 2016 to August 2022. All items of EEG background analysis were enrolled into an assessment system and were scored according to severity to obtain the total EEG score. The correlations of total EEG score with total MRI score and total Sarnat score (TSS, used to evaluate clinical gradings) were analyzed by Spearman correlation analysis. The total EEG score was compared among the neonates with different clinical gradings and among the neonates with different head MRI gradings. The receiver operating characteristic (ROC) curve and the area under thecurve (AUC) were used to evaluate the value of total EEG score in diagnosing moderate/severe head MRI abnormalities and clinical moderate/severe HIE, which was then compared with the aEEG grading method. RESULTS A total of 50 neonates with HIE were included. The total EEG score was positively correlated with the total head MRI score and TSS (rs=0.840 and 0.611 respectively, P<0.001). There were significant differences in the total EEG score between different clinical grading groups and different head MRI grading groups (P<0.05). The total EEG score and the aEEG grading method had an AUC of 0.936 and 0.617 respectively in judging moderate/severe head MRI abnormalities (P<0.01) and an AUC of 0.887 and 0.796 respectively in judging clinical moderate/severe HIE (P>0.05). The total EEG scores of ≤6 points, 7-13 points, and ≥14 points were defined as mild, moderate, and severe EEG abnormalities respectively, which had the best consistency with clinical grading and head MRI grading (P<0.05). CONCLUSIONS The new EEG background scoring method can quantitatively reflect the severity of brain injury and can be used for the judgment of brain function in neonates with HIE.
Collapse
Affiliation(s)
- Xiu-Ying Fang
- Department of Neonatology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yi-Li Tian
- Department of Neonatology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shu-Yuan Chen
- Department of Neonatology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Quan Shi
- Department of Neonatology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Duo Zheng
- Department of Neonatology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | | | | |
Collapse
|
23
|
Bourel-Ponchel E, Querne L, Flamein F, Ghostine-Ramadan G, Wallois F, Lamblin MD. The prognostic value of neonatal conventional-EEG monitoring in hypoxic-ischemic encephalopathy during therapeutic hypothermia. Dev Med Child Neurol 2023; 65:58-66. [PMID: 35711160 PMCID: PMC10084260 DOI: 10.1111/dmcn.15302] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 01/28/2023]
Abstract
AIM To determine the prognostic value of conventional electroencephalography (EEG) monitoring in neonatal hypoxic-ischemic encephalopathy (HIE). METHOD In this multicentre retrospective study, 95 full-term neonates (mean of 39.3wks gestational age [SD 1.4], 36 [38%] females, 59 [62%] males) with HIE (2013-2016) undergoing therapeutic hypothermia were divided between favourable or adverse outcomes. Background EEG activity (French classification scale: 0-1-2-3-4-5) and epileptic seizure burden (epileptic seizure scale: 0-1-2) were graded for seven 6-hour periods. Conventional EEG monitoring was investigated by principal component analysis (PCA), with clustering methods to extract prognostic biomarkers of development at 2 years and infant death. RESULTS Eighty-one per cent of infants with an adverse outcome had a French classification scale equal to or greater than 3 after H48 (100% at H6-12). The H6-12 epileptic seizure scale was equal to or greater than 1 for 39%, increased to 52% at H30-36 and then remained equal to or greater than 1 for 39% after H48. Forty-five per cent of infants with a favourable outcome had a H6-12 French classification scale equal to or greater than 3, which dropped to 5% after H48; 13% had a H6-12 epileptic seizure scale equal to or greater than 1 but no seizures after H48. Clustering methods based on PCA showed the high efficiency (96%) of conventional EEG monitoring for outcome prediction and allowed the definition of three prognostic EEG biomarkers: H6-78 French classification scale mean, H6-78 French classification scale slope, and H30-78 epileptic seizure scale mean. INTERPRETATION Early lability and recovery of physiological features is prognostic of a favourable outcome. Seizure onset from the second day should also be considered to accurately predict neurodevelopment in HIE and support the importance of conventional EEG monitoring in HIE in infants cooled with therapeutic hypothermia. WHAT THIS PAPER ADDS Comprehensive analysis showed the high prognostic efficiency (96%) of conventional electroencephalography (EEG) monitoring. Prognostic EEG biomarkers consist of the grade of background EEG activity, its evolution, and the mean seizure burden. Persistent seizures (H48) without an improvement in background EEG activity were consistently associated with an adverse outcome.
Collapse
Affiliation(s)
- Emilie Bourel-Ponchel
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, Amiens, France.,Pediatric Neurophysiology Unit, Amiens Picardie University Hospital, Amiens, France
| | - Laurent Querne
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, Amiens, France.,Department of Pediatric Neurology, Amiens-Picardie University Hospital, Amiens, France
| | - Florence Flamein
- Department of Neonatology, University Hospital of Lille, Lille, France
| | - Ghida Ghostine-Ramadan
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, Amiens, France.,Neonatal Intensive Care Unit, Amiens-Picardie University Hospital, Amiens, France
| | - Fabrice Wallois
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, Amiens, France.,Pediatric Neurophysiology Unit, Amiens Picardie University Hospital, Amiens, France
| | | |
Collapse
|
24
|
El-Dib M, Abend NS, Austin T, Boylan G, Chock V, Cilio MR, Greisen G, Hellström-Westas L, Lemmers P, Pellicer A, Pressler RM, Sansevere A, Tsuchida T, Vanhatalo S, Wusthoff CJ, Wintermark P, Aly H, Chang T, Chau V, Glass H, Lemmon M, Massaro A, Wusthoff C, deVeber G, Pardo A, McCaul MC. Neuromonitoring in neonatal critical care part I: neonatal encephalopathy and neonates with possible seizures. Pediatr Res 2022:10.1038/s41390-022-02393-1. [PMID: 36476747 DOI: 10.1038/s41390-022-02393-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/12/2022] [Accepted: 08/19/2022] [Indexed: 12/12/2022]
Abstract
The blooming of neonatal neurocritical care over the last decade reflects substantial advances in neuromonitoring and neuroprotection. The most commonly used brain monitoring tools in the neonatal intensive care unit (NICU) are amplitude integrated EEG (aEEG), full multichannel continuous EEG (cEEG), and near-infrared spectroscopy (NIRS). While some published guidelines address individual tools, there is no consensus on consistent, efficient, and beneficial use of these modalities in common NICU scenarios. This work reviews current evidence to assist decision making for best utilization of neuromonitoring modalities in neonates with encephalopathy or with possible seizures. Neuromonitoring approaches in extremely premature and critically ill neonates are discussed separately in the companion paper. IMPACT: Neuromonitoring techniques hold promise for improving neonatal care. For neonatal encephalopathy, aEEG can assist in screening for eligibility for therapeutic hypothermia, though should not be used to exclude otherwise eligible neonates. Continuous cEEG, aEEG and NIRS through rewarming can assist in prognostication. For neonates with possible seizures, cEEG is the gold standard for detection and diagnosis. If not available, aEEG as a screening tool is superior to clinical assessment alone. The use of seizure detection algorithms can help with timely seizures detection at the bedside.
Collapse
Affiliation(s)
- Mohamed El-Dib
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Nicholas S Abend
- Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - Topun Austin
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Geraldine Boylan
- INFANT Research Centre & Department of Paediatrics & Child Health, University College Cork, Cork, Ireland
| | - Valerie Chock
- Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - M Roberta Cilio
- Department of Pediatrics, Division of Pediatric Neurology, Cliniques universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Gorm Greisen
- Department of Neonatology, Rigshospitalet, Copenhagen University Hospital & Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lena Hellström-Westas
- Department of Women's and Children's Health, Uppsala University, and Division of Neonatology, Uppsala University Hospital, Uppsala, Sweden
| | - Petra Lemmers
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Adelina Pellicer
- Department of Neonatology, La Paz University Hospital, Madrid, Spain; Neonatology Group, IdiPAZ, Madrid, Spain
| | - Ronit M Pressler
- Department of Clinical Neurophysiology, Great Ormond Street Hospital for Children NHS Trust, and Clinical Neuroscience, UCL- Great Ormond Street Institute of Child Health, London, UK
| | - Arnold Sansevere
- Department of Neurology and Pediatrics, George Washington University School of Medicine and Health Sciences; Children's National Hospital Division of Neurophysiology, Epilepsy and Critical Care, Washington, DC, USA
| | - Tammy Tsuchida
- Department of Neurology and Pediatrics, George Washington University School of Medicine and Health Sciences; Children's National Hospital Division of Neurophysiology, Epilepsy and Critical Care, Washington, DC, USA
| | - Sampsa Vanhatalo
- Department of Clinical Neurophysiology, Children's Hospital, BABA Center, Neuroscience Center/HILIFE, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Johnson KJ, Moy B, Rensing N, Robinson A, Ly M, Chengalvala R, Wong M, Galindo R. Functional neuropathology of neonatal hypoxia-ischemia by single-mouse longitudinal electroencephalography. Epilepsia 2022; 63:3037-3050. [PMID: 36054439 PMCID: PMC10176800 DOI: 10.1111/epi.17403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Neonatal cerebral hypoxia-ischemia (HI) results in symptomatic seizures and long-term neurodevelopmental disability. The Rice-Vannucci model of rodent neonatal HI has been used extensively to examine and translate the functional consequences of acute and chronic HI-induced encephalopathy. Yet, longitudinal electrophysiological characterization of this brain injury model has been limited by the size of the neonatal mouse's head and postnatal maternal dependency. We overcome this challenge by employing a novel method of longitudinal single-mouse electroencephalography (EEG) using chronically implanted subcranial electrodes in the term-equivalent mouse pup. We characterize the neurophysiological disturbances occurring during awake and sleep states in the acute and chronic phases following newborn brain injury. METHODS C57BL/6 mice underwent long-term bilateral subcranial EEG and electromyographic electrode placement at postnatal day 9 followed by unilateral carotid cauterization and exposure to 40 minutes of hypoxia the following day. EEG recordings were obtained prior, during, and intermittently after the HI procedure from postnatal day 10 to weaning age. Quantitative EEG and fast Fourier transform analysis were used to evaluate seizures, cortical cerebral dysfunction, and disturbances in vigilance states. RESULTS We observed neonatal HI-provoked electrographic focal and bilateral seizures during or immediately following global hypoxia and most commonly contralateral to the ischemic injury. Spontaneous chronic seizures were not seen. Injured mice developed long-term asymmetric EEG background attenuation in all frequencies and most prominently during non-rapid eye movement (NREM) sleep. HI mice also showed transient impairments in vigilance state duration and transitions during the first 2 days following injury. SIGNIFICANCE The functional burden of mouse neonatal HI recorded by EEG resembles closely that of the injured human newborn. The use of single-mouse longitudinal EEG in this immature model can advance our understanding of the developmental and pathophysiological mechanisms of neonatal cerebral injury and help translate novel therapeutic strategies against this devastating condition.
Collapse
Affiliation(s)
- Kevin J Johnson
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brianna Moy
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicholas Rensing
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alexia Robinson
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael Ly
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ramya Chengalvala
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael Wong
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rafael Galindo
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
26
|
Starodubtseva NL, Eldarov C, Kirtbaya AR, Balashova EN, Gryzunova AS, Ionov OV, Zubkov VV, Silachev DN. Recent advances in diagnostics of neonatal hypoxic ischemic encephalopathy. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2022. [DOI: 10.24075/brsmu.2022.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The prognosis in neonatal hypoxic ischemic encephalopathy (HIE) depends on early differential diagnosis for justified administration of emergency therapeutic hypothermia. The moment of therapy initiation directly affects the long-term neurological outcome: the earlier the commencement, the better the prognosis. This review analyzes recent advances in systems biology that facilitate early differential diagnosis of HIE as a pivotal complement to clinical indicators. We discuss the possibilities of clinical translation for proteomic, metabolomic and extracellular vesicle patterns characteristic of HIE and correlations with severity and prognosis. Identification and use of selective biomarkers of brain damage in neonates during the first hours of life is hindered by systemic effects of hypoxia. Chromatography– mass spectrometry blood tests allow analyzing hundreds and thousands of metabolites in a small biological sample to identify characteristic signatures of brain damage. Clinical use of advanced analytical techniques will facilitate the accurate and timely diagnosis of HIE for enhanced management.
Collapse
Affiliation(s)
- NL Starodubtseva
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - ChM Eldarov
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - AR Kirtbaya
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - EN Balashova
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - AS Gryzunova
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - OV Ionov
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - VV Zubkov
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - DN Silachev
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| |
Collapse
|
27
|
Troha Gergeli A, Škofljanec A, Neubauer D, Paro Panjan D, Kodrič J, Osredkar D. Prognostic Value of Various Diagnostic Methods for Long-Term Outcome of Newborns After Hypoxic-Ischemic Encephalopathy Treated With Hypothermia. Front Pediatr 2022; 10:856615. [PMID: 35463898 PMCID: PMC9021608 DOI: 10.3389/fped.2022.856615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Prediction of outcome in newborns with hypoxic-ischemic encephalopathy (HIE) has been modulated by hypothermia treatment (HT). We assessed the predictive value of diagnostic methods commonly used in neonates with HIE for short-term neurodevelopmental outcome and long-term neurological outcome. MATERIALS AND METHODS This longitudinal cohort study followed up 50 term newborns who underwent HT after HIE between July 2006 and August 2015, until preschool age. We estimated sensitivity and specificity for short-term neurodevelopmental outcome at 18 months and long-term neurological outcome at five years based on Amiel-Tison Neurological Assessment (ATNA), electroencephalography (EEG), and magnetic resonance imaging (MRI) performed in the neonatal period. RESULTS The accuracy of all neonatal methods tested was higher for long-term neurological outcome compared to the predictive accuracy for short-term neurodevelopmental outcome at 18-24 months. Sensitivity and specificity in predicting unfavorable long-term neurological outcome were: MRI (sensitivity 1.0 [95%CI 0.96-1.0]; specificity 0.91 [95%CI 0.86-1.0]), EEG (sensitivity 0.94 [95%CI 0.71-1.0]; specificity 1.0 [95% CI 0.89-1.0]), and ATNA (sensitivity 0.94 [95%CI 0.71-1.0]; specificity 0.91 [95%CI 0.76-0.98]). CONCLUSION MRI is a powerful predictor of long-term neurological outcome when performed in the first week after HIE in HT treated infants, as are EEG and ATNA performed in the second or third week postnatally.
Collapse
Affiliation(s)
- Anja Troha Gergeli
- Department of Child, Adolescent and Developmental Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Andreja Škofljanec
- Pediatric Intensive Care, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Health Institution Zdravje, Ljubljana, Slovenia
| | - David Neubauer
- Department of Child, Adolescent and Developmental Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Center for Developmental Neuroscience, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Darja Paro Panjan
- Center for Developmental Neuroscience, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Neonatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Jana Kodrič
- Unit of Child Psychiatry of the University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Damjan Osredkar
- Department of Child, Adolescent and Developmental Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Center for Developmental Neuroscience, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
28
|
Wainwright MS, Guilliams K, Kannan S, Simon DW, Tasker RC, Traube C, Pineda J. Acute Neurologic Dysfunction in Critically Ill Children: The PODIUM Consensus Conference. Pediatrics 2022; 149:S32-S38. [PMID: 34970681 DOI: 10.1542/peds.2021-052888e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 11/24/2022] Open
Abstract
CONTEXT Acute neurologic dysfunction is common in critically ill children and contributes to outcomes and end of life decision-making. OBJECTIVE To develop consensus criteria for neurologic dysfunction in critically ill children by evaluating the evidence supporting such criteria and their association with outcomes. DATA SOURCES Electronic searches of PubMed and Embase were conducted from January 1992 to January 2020, by using a combination of medical subject heading terms and text words to define concepts of neurologic dysfunction, pediatric critical illness, and outcomes of interest. STUDY SELECTION Studies were included if the researchers evaluated critically ill children with neurologic injury, evaluated the performance characteristics of assessment and scoring tools to screen for neurologic dysfunction, and assessed outcomes related to mortality, functional status, organ-specific outcomes, or other patient-centered outcomes. Studies with an adult population or premature infants (≤36 weeks' gestational age), animal studies, reviews or commentaries, case series with sample size ≤10, and studies not published in English with an inability to determine eligibility criteria were excluded. DATA EXTRACTION Data were abstracted from each study meeting inclusion criteria into a standard data extraction form by task force members. DATA SYNTHESIS The systematic review supported the following criteria for neurologic dysfunction as any 1 of the following: (1) Glasgow Coma Scale score ≤8; (2) Glasgow Coma Scale motor score ≤4; (3) Cornell Assessment of Pediatric Delirium score ≥9; or (4) electroencephalography revealing attenuation, suppression, or electrographic seizures. CONCLUSIONS We present consensus criteria for neurologic dysfunction in critically ill children.
Collapse
Affiliation(s)
- Mark S Wainwright
- Division of Pediatric Neurology, Department of Neurology, School of Medicine, University of Washington, Seattle, Washington
| | - Kristin Guilliams
- Division of Pediatric and Development Neurology, Department of Neurology and Division of Pediatric Critical Care Medicine, Department of Pediatrics, School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Dennis W Simon
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert C Tasker
- Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Chani Traube
- Division of Critical Care Medicine, Department of Pediatrics, Weill Cornell Medical College, New York
| | - Jose Pineda
- Department of Anesthesiology Critical Care, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California
| | | |
Collapse
|
29
|
Sandoval Karamian AG, Wusthoff CJ. Current and Future Uses of Continuous EEG in the NICU. Front Pediatr 2021; 9:768670. [PMID: 34805053 PMCID: PMC8595393 DOI: 10.3389/fped.2021.768670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/12/2021] [Indexed: 11/28/2022] Open
Abstract
Continuous EEG (cEEG) is a fundamental neurodiagnostic tool in the care of critically ill neonates and is increasingly recommended. cEEG enhances prognostication via assessment of the background brain activity, plays a role in predicting which neonates are at risk for seizures when combined with clinical factors, and allows for accurate diagnosis and management of neonatal seizures. Continuous EEG is the gold standard method for diagnosis of neonatal seizures and should be used for detection of seizures in high-risk clinical conditions, differential diagnosis of paroxysmal events, and assessment of response to treatment. High costs associated with cEEG are a limiting factor in its widespread implementation. Centralized remote cEEG interpretation, automated seizure detection, and pre-natal EEG are potential future applications of this neurodiagnostic tool.
Collapse
Affiliation(s)
| | - Courtney J. Wusthoff
- Division of Child Neurology, Lucile Packard Children's Hospital at Stanford, Palo Alto, CA, United States
| |
Collapse
|
30
|
An Introduction to Neonatal EEG. J Perinat Neonatal Nurs 2021; 35:369-376. [PMID: 34726654 DOI: 10.1097/jpn.0000000000000599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Newborn care has witnessed significant improvements in survival, but ongoing concerns persist about neurodevelopmental outcome. Protecting the newborn brain is the focus of neurocritical care in the intensive care unit. Brain-focused care places emphasis on clinical practices supporting neurodevelopment in conjunction with early detection, diagnosis, and treatment of brain injury. Technology now facilitates continuous cot-side monitoring of brain function. Neuromonitoring techniques in neonatal intensive care units include the use of electroencephalography (EEG) or amplitude-integrated EEG (aEEG) and near-infrared spectroscopy. This article aims to provide an introduction to EEG, which is appropriate for neonatal healthcare professionals.
Collapse
|
31
|
Chalak L, Hellstrom-Westas L, Bonifacio S, Tsuchida T, Chock V, El-Dib M, Massaro AN, Garcia-Alix A. Bedside and laboratory neuromonitoring in neonatal encephalopathy. Semin Fetal Neonatal Med 2021; 26:101273. [PMID: 34393094 PMCID: PMC8627431 DOI: 10.1016/j.siny.2021.101273] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Several bedside and laboratory neuromonitoring tools are currently used in neonatal encephalopathy (NE) to assess 1) brain function [amplitude-integrated electroencephalogram (aEEG) and EEG], 2) cerebral oxygenation delivery and consumption [near-infrared spectroscopy (NIRS)] and 3) blood and cerebrospinal fluid biomarkers. The aim of the review is to provide the role of neuromonitoring in understanding the development of brain injury in these newborns and better predict their long-term outcome. Simultaneous use of these monitoring modalities may improve our ability to provide meaningful prognostic information regarding ongoing treatments. Evidence will be summarized in this review for each of these modalities, by describing (1) the methods, (2) the clinical evidence in context of NE both before and with hypothermia, and (3) the research and future directions.
Collapse
Affiliation(s)
- L Chalak
- University of Texas Southwestern Medical Center, Dallas, USA.
| | - L Hellstrom-Westas
- Department of Women's and Children's Health, Uppsala University, Division of Neonatology, Uppsala University Hospital, Sweden.
| | - S Bonifacio
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine; 750 Welch Road, Suite 315, Palo Alto, CA, 94304, USA.
| | - T Tsuchida
- Department of Neurology and Pediatrics, George Washington University School of Medicine and Health Sciences, Children's National Hospital Division of Neurophysiology, Epilepsy and Critical Care, 111 Michigan Ave NW, West Wing, 4th Floor, Washington DC, 20010-2970, USA.
| | - V Chock
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine; 750 Welch Road, Suite 315, Palo Alto, CA, 94304, USA.
| | - M El-Dib
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, CWN#418, Boston, MA, 02115, USA.
| | - AN Massaro
- Department of Pediatrics, The George Washington University School of Medicine and Division of Neonatology, Children’s National Hospital, Washington, USA
| | - A Garcia-Alix
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona, Spain; University of Barcelona, Barcelona, Spain; NeNe Foundation, Madrid, Spain; Passeig de Sant Joan de Déu, 2, 08950, Esplugues de Llobregat, Barcelona, Spain.
| | | |
Collapse
|
32
|
白 文, 方 秀, 石 权, 田 艺, 郑 铎, 陈 淑, 王 英, 毛 健. Correlation of electroencephalogram background evolution with the degree of brain injury in neonates with hypoxic-ischemic encephalopathy. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:909-915. [PMID: 34535205 PMCID: PMC8480165 DOI: 10.7499/j.issn.1008-8830.2105054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/16/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To study the correlation of electroencephalogram (EEG) background evolution with the degree of brain injury in neonates with hypoxic-ischemic encephalopathy (HIE). METHODS A retrospective analysis was performed for 56 neonates with HIE who underwent continuous video electroencephalogram (cVEEG) and brain magnetic resonance imaging (MRI) examinations. According to clinical symptoms, they were divided into a mild group with 3 neonates, a moderate group with 36 neonates, and a severe group with 17 neonates. EEG background grading and MRI score were determined for each group to analyze the correlation of EEG background evolution with the degree of brain injury. RESULTS Compared with the moderate group, the severe group had significantly lower gestational age and Apgar score at 5 minutes after birth, a significantly higher resuscitation score, significantly lower base excess in umbilical cord blood or blood gas within 1 hour, a significantly higher proportion of neonates on mechanical ventilation, and a significantly higher incidence rate of short-term adverse outcomes (P<0.05). For the neonates in the mild and moderate groups, MRI mainly showed no brain injury (67%, 2/3) and watershed injury (67%, 16/24) respectively, and EEG showed mild abnormality in 62% (13/21) of the neonates on the 3rd day after birth. For the neonates in the severe group, MRI mainly showed basal ganglia/thalamus + brainstem injury (24%, 4/17) and whole brain injury (71%, 12/17), and EEG showed moderate or severe abnormalities on the 3rd day after birth. EEG background grading was correlated with clinical grading, MRI score, and short-term outcome on days 1, 2, 3 and 7-14 after birth (P<0.01). The highest correlation coefficient between EEG grading and MRI score was observed on the 3rd day after birth (rs=0.751, P<0.001), and the highest correlation coefficients between EEG grading and clinical grading (rs=0.592, P=0.002) and between EEG grading and short-term outcome (rs=0.737, P<0.001) were observed 7-14 days after birth. Among the neonates with severe abnormal EEG, the neonates without brain electrical activity had the highest MRI score, followed by those with status epileptics and persistent low voltage (P<0.05). CONCLUSIONS There is a good correlation between EEG background grading and degree of brain injury in neonates with HIE, which can help to evaluate the degree and prognosis of brain injury in the early stage.
Collapse
Affiliation(s)
| | - 秀英 方
- 中国医科大学附属盛京医院,神经功能科,辽宁沈阳110004
| | - 权 石
- 中国医科大学附属盛京医院,神经功能科,辽宁沈阳110004
| | - 艺丽 田
- 中国医科大学附属盛京医院,神经功能科,辽宁沈阳110004
| | - 铎 郑
- 中国医科大学附属盛京医院,神经功能科,辽宁沈阳110004
| | - 淑媛 陈
- 中国医科大学附属盛京医院,神经功能科,辽宁沈阳110004
| | | | | |
Collapse
|
33
|
Trollmann R. Neuromonitoring bei zerebralen Anfällen im Neugeborenenalter – Chancen und Herausforderungen. KLIN NEUROPHYSIOL 2021. [DOI: 10.1055/a-1438-1828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Angesichts der vielen Kinder mit neonatalen Risikofaktoren für erworbene ZNS-Läsionen und zerebrale Anfälle ist das EEG zunehmend relevant für eine optimierte Diagnostik und Therapieüberwachung 1
2. Folgender Artikel gibt einen Überblick über Besonderheiten des neonatalen EEG und über aktuelle Empfehlungen zum Stellenwert des Langzeit-EEG-Monitorings bei neonatalen Anfällen und epileptischen Enzephalopathien im Früh- und Neugeborenalter.
Collapse
|
34
|
Wood TR, Vu PT, Comstock BA, Law JB, Mayock DE, Heagerty PJ, Burbacher T, Bammler TK, Juul SE. Cytokine and chemokine responses to injury and treatment in a nonhuman primate model of hypoxic-ischemic encephalopathy treated with hypothermia and erythropoietin. J Cereb Blood Flow Metab 2021; 41:2054-2066. [PMID: 33554708 PMCID: PMC8327104 DOI: 10.1177/0271678x21991439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Predicting long-term outcome in infants with hypoxic-ischemic encephalopathy (HIE) remains an ongoing clinical challenge. We investigated plasma biomarkers and their association with 6-month outcomes in a nonhuman primate model of HIE with or without therapeutic hypothermia (TH) and erythropoietin (Epo). Twenty-nine Macaca nemestrina were randomized to control cesarean section (n = 7) or 20 min of umbilical cord occlusion (UCO, n = 22) with either no treatment (n = 11) or TH/Epo (n = 11). Initial injury severity was scored using 30-min arterial pH, base deficit, and 10-min Apgar score. Twenty-four plasma cytokines, chemokines, and growth factors were measured 3, 6, 24, 72, and 96 h after UCO. Interleukin 17 (IL-17) and macrophage-derived chemokine (MDC) differentiated the normal/mild from moderate/severe injury groups. Treatment with TH/Epo was associated with increased monocyte chemotactic protein-4 (MCP-4) at 3 h-6h, and significantly lower MCP-4 and MDC at 24 h-72h, respectively. IL-12p40 was lower at 24 h-72h in animals with death/cerebral palsy (CP) compared to survivors without CP. Baseline injury severity was the single best predictor of death/CP, and predictions did not improve with the addition of biomarker data. Circulating chemokines associated with the peripheral monocyte cell lineage are associated with severity of injury and response to therapy, but do not improve ability to predict outcomes.
Collapse
Affiliation(s)
- Thomas R Wood
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Phuong T Vu
- Department of Biostatistics, University of Washington, Seattle, WA, USA.,Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Bryan A Comstock
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Janessa B Law
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Dennis E Mayock
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Thomas Burbacher
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Sandra E Juul
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
35
|
Selvanathan T, Miller SP. Early EEG in neonates with mild hypoxic-ischemic encephalopathy: more than meets the eye. Pediatr Res 2021; 90:18-19. [PMID: 33824445 DOI: 10.1038/s41390-021-01514-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Thiviya Selvanathan
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Steven P Miller
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
36
|
Ambalavanan N, Shankaran S, Laptook AR, Carper BA, Das A, Carlo WA, Cotten CM, Duncan AF, Higgins RD. Early Determination of Prognosis in Neonatal Moderate or Severe Hypoxic-Ischemic Encephalopathy. Pediatrics 2021; 147:peds.2020-048678. [PMID: 33986149 PMCID: PMC8168606 DOI: 10.1542/peds.2020-048678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Early determination of prognosis is important in neonates with hypoxic-ischemic encephalopathy (HIE). Our objective was to test scoring systems developed earlier (original scoring system) and develop new prognostic models. METHODS Secondary analysis of data from the multicenter randomized controlled trial of longer, deeper, or usual care cooling in neonatal HIE (NCT01192776) that enrolled 364 neonates diagnosed with moderate or severe HIE. The primary outcome was death or moderate or severe disability at 18 to 22 months, and secondary outcome was death during initial hospitalization. Testing of early neurologic clinical examination (<6 hours of age) and the original scoring system for prognostic ability was done, followed by development of new scoring systems and classification and regression tree (CART) models by using early clinical variables (<6 hours of age). RESULTS For death or disability, the original scoring system correctly classified 75% (95% confidence interval: 69%-81%), whereas the new scoring system correctly classified 78% (73%-82%), and the CART model correctly classified 76% (72%-81%). Early neurologic clinical examination also had a correct classification rate of 76% (71%-80%). Depth and duration of cooling did not affect prediction. Only a few components of the early neurologic examination were associated with poor outcome. For death, the original scoring system correctly classified 72% (66%-77%), the new scoring system 68% (63%-72%), the new CART model 87% (83%-90%), and early neurologic evaluation 81% (77%-85%). CONCLUSIONS The 3 models (scoring system, CART, and early neurologic evaluation) were comparable in predicting death or disability. For in-hospital death, CART models were superior to scoring systems and early neurologic examination.
Collapse
Affiliation(s)
- Namasivayam Ambalavanan
- Department of Pediatrics, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama;
| | - Seetha Shankaran
- Department of Pediatrics, School of Medicine, Wayne State University, Detroit, Michigan
| | - Abbot R. Laptook
- Department of Pediatrics, Women and Infants Hospital, Providence, Rhode Island
| | - Benjamin A. Carper
- Biostatistics and Epidemiology Division, Research Triangle Institute International, Research Triangle Park, North Carolina
| | - Abhik Das
- Biostatistics and Epidemiology Division, Research Triangle Institute International, Rockville, Maryland
| | - Waldemar A. Carlo
- Department of Pediatrics, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - C. Michael Cotten
- Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina
| | - Andrea F. Duncan
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Rosemary D. Higgins
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland; and,College of Health and Human Services, George Mason University, Fairfax, Virginia
| | | |
Collapse
|
37
|
Tataranno ML, Vijlbrief DC, Dudink J, Benders MJNL. Precision Medicine in Neonates: A Tailored Approach to Neonatal Brain Injury. Front Pediatr 2021; 9:634092. [PMID: 34095022 PMCID: PMC8171663 DOI: 10.3389/fped.2021.634092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/14/2021] [Indexed: 11/27/2022] Open
Abstract
Despite advances in neonatal care to prevent neonatal brain injury and neurodevelopmental impairment, predicting long-term outcome in neonates at risk for brain injury remains difficult. Early prognosis is currently based on cranial ultrasound (CUS), MRI, EEG, NIRS, and/or general movements assessed at specific ages, and predicting outcome in an individual (precision medicine) is not yet possible. New algorithms based on large databases and machine learning applied to clinical, neuromonitoring, and neuroimaging data and genetic analysis and assays measuring multiple biomarkers (omics) can fulfill the needs of modern neonatology. A synergy of all these techniques and the use of automatic quantitative analysis might give clinicians the possibility to provide patient-targeted decision-making for individualized diagnosis, therapy, and outcome prediction. This review will first focus on common neonatal neurological diseases, associated risk factors, and most common treatments. After that, we will discuss how precision medicine and machine learning (ML) approaches could change the future of prediction and prognosis in this field.
Collapse
Affiliation(s)
| | | | | | - Manon J. N. L. Benders
- Department of Neonatology, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
38
|
Abstract
Seizures are the most common neurological emergency in the neonates, and this age group has the highest incidence of seizures compared with any other period of life. The author provides a narrative review of recent advances in the genetics of neonatal epilepsies, new neonatal seizure classification system, diagnostics, and treatment of neonatal seizures based on a comprehensive literature review (MEDLINE using PubMED and OvidSP vendors with appropriate keywords to incorporate recent evidence), personal practice, and experience. Knowledge regarding various systemic and postzygotic genetic mutations responsible for neonatal epilepsy has been exploded in recent times, as well as better delineation of clinical phenotypes associated with rare neonatal epilepsies. An International League Against Epilepsy task force on neonatal seizure has proposed a new neonatal seizure classification system and also evaluated the specificity of semiological features related to particular etiology. Although continuous video electroencephalogram (EEG) is the gold standard for monitoring neonatal seizures, amplitude-integrated EEGs have gained significant popularity in resource-limited settings. There is tremendous progress in the automated seizure detection algorithm, including the availability of a fully convolutional neural network using artificial machine learning (deep learning). There is a substantial need for ongoing research and clinical trials to understand optimal medication selection (first line, second line, and third line) for neonatal seizures, treatment duration of antiepileptic drugs after cessation of seizures, and strategies to improve neuromorbidities such as cerebral palsy, epilepsy, and developmental impairments. Although in recent times, levetiracetam use has been significantly increased for neonatal seizures, a multicenter, randomized, blinded, controlled phase IIb trial confirmed the superiority of phenobarbital over levetiracetam in the acute suppression of neonatal seizures. While there is no single best choice available for the management of neonatal seizures, institutional guidelines should be formed based on a consensus of local experts to mitigate wide variability in the treatment and to facilitate early diagnosis and treatment.
Collapse
Affiliation(s)
- Debopam Samanta
- Child Neurology Section, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| |
Collapse
|
39
|
Trollmann R. Neuromonitoring in Neonatal-Onset Epileptic Encephalopathies. Front Neurol 2021; 12:623625. [PMID: 33603712 PMCID: PMC7884638 DOI: 10.3389/fneur.2021.623625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/12/2021] [Indexed: 12/31/2022] Open
Abstract
Considering the wide spectrum of etiologies of neonatal-onset epileptic encephalopathies (EE) and their unfavorable consequences for neurodevelopmental prognoses, neuromonitoring at-risk neonates is increasingly important. EEG is highly sensitive for early identification of electrographic seizures and abnormal background activity. Amplitude-integrated EEG (aEEG) is recommended as a useful bedside monitoring method but as a complementary tool because of methodical limitations. It is of special significance in monitoring neonates with acute symptomatic as well as structural, metabolic and genetic neonatal-onset EE, being at high risk of electrographic-only and prolonged seizures. EEG/aEEG monitoring is established as an adjunctive tool to confirm perinatal hypoxic-ischemic encephalopathy (HIE). In neonates with HIE undergoing therapeutic hypothermia, burst suppression pattern is associated with good outcomes in about 40% of the patients. The prognostic specificity of EEG/aEEG is lower compared to cMRI. As infants with HIE may develop seizures after cessation of hypothermia, recording for at least 24 h after the last seizure is recommended. Progress in the identification of genetic etiology of neonatal EE constantly increases. However, presently, no specific EEG changes indicative of a genetic variant have been characterized, except for individual variants associated with typical EEG patterns (e.g., KCNQ2, KCNT1). Long-term monitoring studies are necessary to define and classify electro-clinical patterns of neonatal-onset EE.
Collapse
Affiliation(s)
- Regina Trollmann
- Department of Pediatrics and Pediatric Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
40
|
Benninger KL, Inder TE, Goodman AM, Cotten CM, Nordli DR, Shah TA, Slaughter JC, Maitre NL. Perspectives from the Society for Pediatric Research. Neonatal encephalopathy clinical trials: developing the future. Pediatr Res 2021; 89:74-84. [PMID: 32221474 PMCID: PMC7529683 DOI: 10.1038/s41390-020-0859-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/27/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022]
Abstract
The next phase of clinical trials in neonatal encephalopathy (NE) focuses on hypothermia adjuvant therapies targeting alternative recovery mechanisms during the process of hypoxic brain injury. Identifying infants eligible for neuroprotective therapies begins with the clinical detection of brain injury and classification of severity. Combining a variety of biomarkers (serum, clinical exam, EEG, movement patterns) with innovative clinical trial design and analyses will help target infants with the most appropriate and timely treatments. The timing of magnetic resonance imaging (MRI) and MR spectroscopy after NE both assists in identifying the acute perinatal nature of the injury (days 3-7) and evaluates the full extent and evolution of the injury (days 10-21). Early, intermediate outcome of neuroprotective interventions may be best defined by the 21-day neuroimaging, with recognition that the full neurodevelopmental trajectory is not yet defined. An initial evaluation of each new therapy at this time point may allow higher-throughput selection of promising therapies for more extensive investigation. Functional recovery can be assessed using a trajectory of neurodevelopmental evaluations targeted to a prespecified and mechanistically derived hypothesis of drug action. As precision medicine revolutionizes healthcare, it should also include the redesign of NE clinical trials to allow safe, efficient, and targeted therapeutics. IMPACT: As precision medicine revolutionizes healthcare, it should also include the redesign of NE clinical trials to allow faster development of safe, effective, and targeted therapeutics. This article provides a multidisciplinary perspective on the future of clinical trials in NE; novel trial design; study management and oversight; biostatistical methods; and a combination of serum, imaging, and neurodevelopmental biomarkers can advance the field and improve outcomes for infants affected by NE. Innovative clinical trial designs, new intermediate trial end points, and a trajectory of neurodevelopmental evaluations targeted to a prespecified and mechanistically derived hypothesis of drug action can help address common challenges in NE clinical trials and allow for faster selection and validation of promising therapies for more extensive investigation.
Collapse
MESH Headings
- Biomarkers/blood
- Biomedical Research/trends
- Brain Diseases/diagnostic imaging
- Brain Diseases/etiology
- Brain Diseases/physiopathology
- Brain Diseases/therapy
- Clinical Trials as Topic
- Consensus
- Delphi Technique
- Diffusion of Innovation
- Forecasting
- Humans
- Infant, Newborn
- Infant, Newborn, Diseases/diagnostic imaging
- Infant, Newborn, Diseases/etiology
- Infant, Newborn, Diseases/physiopathology
- Infant, Newborn, Diseases/therapy
- Neonatology/trends
- Neuroimaging
- Research Design/trends
- Societies, Medical
- Societies, Scientific
- Time Factors
- Treatment Outcome
Collapse
Affiliation(s)
- Kristen L Benninger
- Division of Neonatology and Center for Perinatal Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Amy M Goodman
- Division of Child Neurology, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | | | - Douglas R Nordli
- Section of Child Neurology, Department of Pediatrics, University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Tushar A Shah
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Children's Hospital of The King's Daughters, Eastern Virginia Medical School, Norfolk, VA, USA
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nathalie L Maitre
- Division of Neonatology and Center for Perinatal Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
41
|
Seizures severity during rewarming can predict seizure outcomes of infants with neonatal hypoxic-ischemic encephalopathy following therapeutic hypothermia. Biomed J 2020; 43:285-292. [PMID: 32684487 PMCID: PMC7424086 DOI: 10.1016/j.bj.2020.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 01/14/2023] Open
Abstract
Background The aim of this study was to examine the predictive value of amplitude-integrated electroencephalography (aEEG) on 12-month seizure outcomes of infants with neonatal hypoxic-ischemic encephalopathy (HIE) treated with therapeutic hypothermia. Methods We conducted this retrospective cohort study in a tertiary neonatal intensive care unit between May 2012 and September 2017. Neonates with HIE who received both therapeutic hypothermia (TH) and aEEG were enrolled. Results A total of 23 infants (14 boys, nine girls) with a mean gestational age of 38.9 weeks were enrolled. Fifteen (65%) infants had moderate HIE and eight (35%) had severe HIE according to modified Sarnat staging. The mean aEEG recording time was 107.5 h. Twenty (86.9%) infants had seizure activity during the first 24 h after cooling and 14 (60.8%) had seizure activity during the first 24 h after rewarming. At 12 months, five (21.7%) infants had poor seizure outcomes. Repetitive seizures or status epilepticus pattern during the first 24 h after rewarming, but not the first 24 h after cooling, were associated with the presence of epilepsy at 12 months (p = 0.037). Conclusions We identified a high incidence of electrographic seizures in infants with neonatal HIE treated with therapeutic hypothermia, and post-neonatal epilepsy in the children who survived after HIE. Repetitive seizures or status epilepticus pattern during the first 24 h after rewarming, but not in the first 24 h after cooling, were associated with the presence of epilepsy at 12 months.
Collapse
|
42
|
León-Lozano MZ, Arnaez J, Valls A, Arca G, Agut T, Alarcón A, Garcia-Alix A. Cerebrospinal fluid levels of neuron-specific enolase predict the severity of brain damage in newborns with neonatal hypoxic-ischemic encephalopathy treated with hypothermia. PLoS One 2020; 15:e0234082. [PMID: 32479533 PMCID: PMC7263594 DOI: 10.1371/journal.pone.0234082] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/18/2020] [Indexed: 01/28/2023] Open
Abstract
Objectives To investigate whether cerebrospinal fluid levels of neuron-specific enolase (CSF-NSE) during the first 72 hours correlate with other tools used to assess ongoing brain damage, including clinical grading of hypoxic-ischemic encephalopathy (HIE), abnormal patterns in amplitude integrated electroencephalography (aEEG), and magnetic resonance imaging (MRI), as well as with the neurodevelopmental outcomes at two years of age. Material and methods Prospective observational study performed in two hospitals between 2009 and 2011. Forty-three infants diagnosed with HIE within 6 hours of life were included. HIE was severe in 20 infants, moderate in 12, and mild in 11. Infants with moderate-to-severe HIE received whole-body cooling. Both the HIE cohort and a control group of 59 infants with suspected infection underwent measurement of CSF-NSE concentrations at between 12 and 72 hours after birth. aEEG monitoring was started at admission and brain MRI was performed within the first 2 weeks. Neurodevelopment was assessed at 24 months. Results The HIE group showed higher levels of CSF-NSE than the control group: median 70 ng/ml (29; 205) vs 10.6 ng/ml (7.7; 12.9); p <0.001. Median levels of CSF-NSE in infants with severe, moderate, and mild HIE were 220.5 ng/ml (120.5; 368.8), 45.5 ng/ml (26, 75.3), and 26 ng/ml (18, 33), respectively. CSF-NSE levels correlated were significantly higher in infants with seizures, abnormal aEEG, or abnormal MRI, compared to those without abnormalities. Infants with an adverse outcome showed higher CSF-NSE levels than those with normal findings (p<0.001), and the most accurate CSF-NSE cutoff level for predicting adverse outcome in the whole cohort was 108 ng/ml and 50ng/ml in surviving infants. Conclusions In the era of hypothermia, CSF-NSE concentrations provides valuable information as a clinical surrogate of the severity of hypoxic-ischemic brain damage, and this information may be predictive of abnormal outcome at two years of age.
Collapse
Affiliation(s)
- Marisol-Zulema León-Lozano
- Althaia Xarxa, Assistencial Universitária de Manresa, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Juan Arnaez
- Department of Neonatology, Hospital Universitario de Burgos, Burgos, Spain
- NeNe Foundation, Madrid, Spain
| | - Ana Valls
- Institut de Recerca Sant Joan de Dèu, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Gemma Arca
- NeNe Foundation, Madrid, Spain
- Department of Neonatology, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Thais Agut
- NeNe Foundation, Madrid, Spain
- Department of Neonatology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Ana Alarcón
- Department of Neonatology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Alfredo Garcia-Alix
- University of Barcelona, Barcelona, Spain
- NeNe Foundation, Madrid, Spain
- Institut de Recerca Sant Joan de Dèu, Hospital Sant Joan de Déu, Barcelona, Spain
- CIBER de Enfermedades Raras, Madrid, Spain
- * E-mail:
| |
Collapse
|
43
|
Liu W, Yang Q, Wei H, Dong W, Fan Y, Hua Z. Prognostic Value of Clinical Tests in Neonates With Hypoxic-Ischemic Encephalopathy Treated With Therapeutic Hypothermia: A Systematic Review and Meta-Analysis. Front Neurol 2020; 11:133. [PMID: 32161566 PMCID: PMC7052385 DOI: 10.3389/fneur.2020.00133] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/05/2020] [Indexed: 12/17/2022] Open
Abstract
Background and Objective: There remains an unmet clinical need for markers that predict outcomes in the hypothermia-treated (HT) infants with HIE. The aim of this meta-analysis was to investigate the prognostic accuracy of currently available clinical tests performed in the immediate post-natal period for predicting neurological outcomes between 18 months and 3 years of age in HT near-term and term infants with perinatal asphyxia and HIE. Methods: A comprehensive review of the Embase, Cochrane library, and PubMed databases was performed to identify studies that evaluated the prognostic value of clinical tests for neurological outcomes in HT near-term and term infants with perinatal asphyxia and hypoxic-ischemic encephalopathy. Pooled sensitivity and specificity with corresponding 95% confidence intervals and area under the receiver operating characteristic (ROC) curve (AUC) were calculated. Results: Of the 1,144 relevant studies, 26 studies describing four clinical tests conducted in 1458 HT near-term or term infants were included. For predicting an unfavorable neurological outcome, of the imaging techniques, MRI within 2 weeks of birth performed best on sensitivity 0.85 (95% CI 0.79–0.89), specificity 0.72 (95% CI 0.66–0.77), and AUC 0.88; among the neurophysiological tests, multichannel EEG (Electroencephalogram) demonstrated the sensitivity 0.63 (95% CI 0.49–0.76), specificity 0.82 (95% CI 0.70–0.91), and AUC 0.88, and for aEEG (amplitude-integrated electroencephalography) background pattern pooled sensitivity, specificity and AUC were 0.90 (95% CI 0.86–0.94), 0.46 (95% CI 0.42–0.51), and 0.78 whereas for SEPs (Somatosensory evoked potentials), pooled sensitivity and specificity were 0.52 (95% CI 0.34–0.69), 0.76 (95% CI 0.63–0.87), and AUC 0.84, respectively. Conclusions: In the wake of the era of TH, MRI and neurophysiological tests (aEEG or EEG) were promising predictors of adverse outcomes, while SEPs need high-quality studies to confirm the findings. Continued follow-up of the children and well-designed large prospective studies are essential to determine whether these benefits are maintained in later childhood.
Collapse
Affiliation(s)
- Weiqin Liu
- Department of Neonatology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qifen Yang
- School of Life Sciences, SouthWest University, Chongqing, China
| | - Hong Wei
- Department of Neonatology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wenhui Dong
- Department of Neonatology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Fan
- Department of Neonatology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ziyu Hua
- Department of Neonatology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
44
|
Echeverría-Palacio CM, Agut T, Arnaez J, Valls A, Reyne M, Garcia-Alix A. Neuron-Specific Enolase in Cerebrospinal Fluid Predicts Brain Injury After Sudden Unexpected Postnatal Collapse. Pediatr Neurol 2019; 101:71-77. [PMID: 31023601 DOI: 10.1016/j.pediatrneurol.2019.02.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/13/2019] [Accepted: 02/25/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Biomarkers of brain injury with high predictive value in newborns in critical neurological status are increasingly required. Neuron-specific enolase in cerebrospinal fluid has been shown to be highly predictive in newborns with perinatal hypoxic-ischemic encephalopathy, but its utility has not been examined in sudden unexpected postnatal collapse. PURPOSE We analyzed whether the levels of neuron-specific enolase in cerebrospinal fluid can be a useful biomarker to estimate the severity of brain injury in neonates after a sudden unexpected postnatal collapse. METHODS This is a prospective observational study of near-term infants who were consecutively admitted with sudden unexpected postnatal collapse in two neonatal intensive care units during a nine-year period. Variables were collected and analyzed regarding the perinatal period, clinical course, severity of encephalopathy, amplitude-integrated encephalography, magnetic resonance imaging findings, and outcome. Neuron-specific enolase in cerebrospinal fluid samples were obtained in 18 infants with sudden unexpected postnatal collapse between 12 and 72 hours after the collapse and compared with those of 29 controls. RESULTS The levels of neuron-specific enolase in cerebrospinal fluid were higher in patients than in controls (P < 0.001). Levels of neuron-specific enolase in cerebrospinal fluid in infants with sudden unexpected postnatal collapse were significantly higher in patients who presented severe encephalopathy, seizures, abnormal amplitude-integrated encephalography background, or brain injury on magnetic resonance imaging. Receiver operator characteristic curve analysis revealed a neuron-specific enolase in cerebrospinal fluid cutoff value of maximum predictive accuracy of 61 ng/mL (area under the curve, 1.0; sensitivity, specificity, positive predictive value, and negative predictive value, 100%) for identifying infants who died or had adverse outcomes. CONCLUSIONS Levels of neuron-specific enolase in cerebrospinal fluid obtained between 12 and 72 hours after a sudden unexpected postnatal collapse event seem to be a useful biomarker for identifying newborns with severe brain injury and for predicting outcome.
Collapse
Affiliation(s)
- Carlos Mario Echeverría-Palacio
- Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, Barcelona, Spain; Research Group "Neuroped-UNAL", School of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Thais Agut
- Department of Neonatology, Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Juan Arnaez
- Department of Neonatology, Hospital Universitario de Burgos, Burgos, Spain; Fundación NeNe, Spain
| | - Ana Valls
- Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Mar Reyne
- Department of Neonatology, Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Alfredo Garcia-Alix
- Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, Barcelona, Spain; Universitat de Barcelona, Barcelona, Spain; CIBERER. U724, Madrid, Spain; Fundación NeNe, Spain.
| |
Collapse
|
45
|
Dwivedi D, Lin N, Venkatesan C, Kline-Fath B, Holland K, Schapiro M. Clinical, Neuroimaging, and Electrographic Predictors of Phenobarbital Failure in Newborns With Hypoxic Ischemic Encephalopathy and Seizures. J Child Neurol 2019; 34:458-463. [PMID: 30966848 DOI: 10.1177/0883073819838171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Many neonates with hypoxic ischemic encephalopathy and seizures do not respond to the first line antiepileptic drug, phenobarbital. Little is known about what factors are associated with its failure. OBJECTIVE To examine factors associated with failure of phenobarbital therapy in neonates with hypoxic ischemic encephalopathy and seizures. DESIGN/METHODS A single-center retrospective review of 50 term (>35 weeks) neonates with hypoxic ischemic encephalopathy and seizures treated with phenobarbital as the first-line antiepileptic. Neonates were classified into either responders (n = 30) or nonresponders (n = 20). Nonresponse was defined as continued seizures after maximum dosing of phenobarbital or an additional antiepileptic. Subjects with acceptable magnetic resonance imaging (MRI) scans obtained within 2 weeks of birth were included in the study and rated using an MRI injury scoring system. Charts were reviewed for demographic, clinical, and laboratory variables. Resuscitation and seizure scores were also calculated. Electroencephalographic (EEG) background activity was reviewed in 2 different time epochs (12-24 hours and 24-36 hours of life) and graded as per ACNS guidelines. RESULTS There were no significant group differences in demographic, clinical, and laboratory variables except nonresponders, who had higher mean seizure score (P = .01) and significantly more injury on MRI scan for white matter (P = .004), parenchymal cortex (P = .027), and watershed (P = .009) regions. Neonates with moderately abnormal or severely abnormal background EEG responded poorly to phenobarbital. CONCLUSION In the presence of above factors, one can anticipate that additional antiepileptic medication may be needed. These data also support that larger studies should be done to look prospectively at using alternative agents first line in patients with severe injury.
Collapse
Affiliation(s)
- Deepak Dwivedi
- 1 Department of Paediatrics, SS Medical College, Rewa, Madhya Pradesh, India
| | - Nan Lin
- 2 Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Charu Venkatesan
- 2 Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - B Kline-Fath
- 3 Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Katherine Holland
- 2 Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mark Schapiro
- 2 Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,4 Department of Paediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| |
Collapse
|
46
|
Shany E, Taha N, Benkovich E, Novoa R, Meledin I, Mandola A, Novack V, Shelef I. Association of cerebral activity with MRI scans in infants with neonatal encephalopathy undergoing therapeutic hypothermia. Eur J Pediatr 2019; 178:851-861. [PMID: 30900074 DOI: 10.1007/s00431-019-03364-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/01/2019] [Accepted: 02/06/2019] [Indexed: 11/29/2022]
Abstract
We aimed to correlate amplitude-integrated EEG (aEEG) in neonatal hypoxic ischemic encephalopathy (HIE) with early magnetic resonance imaging (MRI). In this retrospective study, 32 neonates over 35 weeks' gestation with moderate/severe HIE who were treated with hypothermia were included. Early MRI scans and daily aEEG background were categorized to mild/normal, moderate, and severely abnormal. Time to sleep cycling was noted on aEEG. Mantel-Haenszel test for trends was used to explore associations between aEEG and MRI and outcome. LOESS regression was used for exploring the association of cycling with MRI scores. MRI was normal/mildly abnormal in 20 (63%) infants; in 9 (28%), moderately abnormal; and in 3 (9%), severely abnormal. Twenty-seven (84%) infants s urvived. MRI severity score was significantly associated with aEEG background score on the third and fourth days of life (p < 0.01). An increase in the MRI severity score was noted if sleep cycling appeared after the fifth day of life.Conclusions: Depressed aEEG at the third and fourth days of life and appearance of cycling beyond the fifth day of life are associated with cerebral MRI abnormalities and may be associated with increased risk of abnormal outcome. What is known: • Since therapeutic hypothermia has been shown to change long-term outcome, amplitude-integrated EEG in infants with hypoxic ischemic neonatal encephalopathy soon after birth have a limited predictive power for long-term outcome in treated infants. • Brain MRI after therapeutic hypothermia in the above infants has a significant predictive value for long-term outcome What is new: • Background amplitude-integrated EEG activity depression at the age of 3 and 4 days and delay of appearance of cycling activity are associated with worse MRI scores and may be predictive of worse long-term outcome.
Collapse
Affiliation(s)
- Eilon Shany
- Neonatology Department, Soroka Medical Center, Rager Ave 151, 84101, Be'er Sheva, Israel. .,Faculty of Health sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel.
| | - Nasrin Taha
- Pediatrics Division, Soroka Medical Center, Be'er Sheva, Israel
| | - Ela Benkovich
- Faculty of Health sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel.,Radiology Department, Soroka Medical Center, Be'er Sheva, Israel
| | - Rosa Novoa
- Faculty of Health sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel.,Radiology Department, Soroka Medical Center, Be'er Sheva, Israel
| | - Irina Meledin
- Neonatology Department, Soroka Medical Center, Rager Ave 151, 84101, Be'er Sheva, Israel.,Faculty of Health sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel
| | - Amarilla Mandola
- Faculty of Health sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel.,Pediatrics Division, Soroka Medical Center, Be'er Sheva, Israel
| | - Victor Novack
- Faculty of Health sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel.,Clinical Research Center, Soroka Medical Center, Be'er Sheva, Israel
| | - Ilan Shelef
- Faculty of Health sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel.,Radiology Department, Soroka Medical Center, Be'er Sheva, Israel
| |
Collapse
|
47
|
Goasdoue K, Chand KK, Miller SM, Lee KM, Colditz PB, Wixey JA, Bjorkman ST. Seizures Are Associated with Blood-Brain Barrier Disruption in a Piglet Model of Neonatal Hypoxic-Ischaemic Encephalopathy. Dev Neurosci 2019; 40:1-16. [PMID: 31048585 DOI: 10.1159/000499365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/04/2019] [Indexed: 11/19/2022] Open
Abstract
Seizures in the neonatal period are most often symptomatic of central nervous system (CNS) dysfunction and the most common cause is hypoxic-ischaemic encephalopathy (HIE). Seizures are associated with poor long-term outcomes and increased neuropathology. Blood-brain barrier (BBB) disruption and inflammation may contribute to seizures and increased neuropathology but are incompletely understood in neonatal HIE. The aim of this study was to investigate the impact of seizures on BBB integrity in a preclinical model of neonatal hypoxic-ischaemic (HI) injury. Piglets (age: <24 h) were subjected to a 30-min HI insult followed by recovery to 72 h post-insult. Amplitude-integrated electroencephalography (aEEG) was performed and seizure burden and background aEEG pattern were analysed. BBB disruption was evaluated in the parietal cortex and hippocampus by means of immunohistochemistry and Western blot. mRNA and protein expression of tight-junction proteins (zonula-occludens 1 [ZO1], occludin [OCLN], and claudin-5 [CLDN5]) was assessed using quantitative polymerase chain reaction (qPCR) and Western blot. In addition, mRNA from genes associated with BBB disruption vascular endothelial growth factor (VEGF) and matrix metalloproteinase 2 (MMP2) as well as inflammatory cytokines and chemokines was assessed with qPCR. Piglets that developed seizures following HI (HI-Sz) had significantly greater injury, as demonstrated by poorer aEEG background pattern scores, lower neurobehavioural scores, and greater histopathology. HI-Sz animals had severe IgG extravasation into brain tissue and uptake into neurons as well as significantly greater levels of IgG in both brain regions as assessed by Western blot. IgG protein in both brain regions was significantly associated with seizure burden, aEEG pattern scores, and neurobehavioural scores. There was no difference in mRNA expression of the tight junctions, however a significant loss of ZO1 and OCLN protein was observed in the parietal cortex. The inflammatory genes TGFβ, IL1β, IL8, IL6, and TNFα were significantly upregulated in HI-Sz animals. MMP2 was significantly increased in animals with seizures compared with animals without seizures. Increasing our understanding of neuropathology associated with seizure is vital because of the association between seizure and poor outcomes. Investigating the BBB is a major untapped area of research and a potential avenue for novel treatments.
Collapse
Affiliation(s)
- Kate Goasdoue
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Kirat Kishore Chand
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Stephanie Melita Miller
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Kah Meng Lee
- Institute of Health Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Queensland, Australia
| | - Paul Bernard Colditz
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Julie Anne Wixey
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Stella Tracey Bjorkman
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia,
| |
Collapse
|
48
|
Han Y, Fu N, Chen W, Liang J, Cui Y, Zhang Y, Qin J. Prognostic Value of Electroencephalography in Hypothermia-Treated Neonates With Hypoxic-Ischemic Encephalopathy: A Meta-Analysis. Pediatr Neurol 2019; 93:3-10. [PMID: 30691779 DOI: 10.1016/j.pediatrneurol.2018.12.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/05/2018] [Accepted: 12/24/2018] [Indexed: 02/01/2023]
Abstract
BACKGROUND Electroencephalography (EEG) background activity is associated with neurological outcome in neonates with hypoxic-ischemic encephalopathy. There is uncertainty about the prognostic value of EEG background activity after hypothermia was introduced. METHODS Searches were made on Pubmed, Embase, and the Cochrane Library, from inception to March 1, 2018. Pooled sensitivities and specificities were calculated to assess the diagnostic power of burst suppression, low voltage, and flat trace background activities in the prediction of an adverse neurological outcome in the follow-up period in hypothermia-treated neonates with hypoxic-ischemic encephalopathy. I2 was used to assess heterogeneity, and meta-regression was done to explore the source of heterogeneity. RESULTS Eighteen studies with 940 neonates were included. Pooled sensitivities and specificities in predicting the combination of death and neurodevelopmental impairment were burst suppression (sensitivity 0.87 [95% confidence interval (CI) 0.79 to 0.93], specificity 0.60 [95% CI 0.44 to 0.74]), low voltage (sensitivity 0.84 [0.75 to 0.90], specificity 0.80 [0.58 to 0.92]), and flat trace (sensitivity 0.85 [0.75 to 0.92], specificity 0.94 [0.77 to 0.99]). Subgroup analysis revealed the sensitivities of background patterns obtained after 24 hours of life were higher than those within age 24 hours, whereas the specificities were just the reverse. Flat trace performed best on sensitivity 0.93 (0.60 to 0.99) and specificity 0.90 (0.64 to 0.98) in predicting death. Burst suppression demonstrated the highest sensitivity 0.87 (0.58 to 0.97) and flat trace performed best on specificity 0.85 (0.60 to 0.96) in predicting neurodevelopmental impairment. CONCLUSIONS EEG background activity is predictive of long-term neurological outcome in hypothermia-treated neonates with hypoxic-ischemic encephalopathy. Burst suppression, low voltage, and flat trace are potential predictors of death or neurodevelopmental impairment.
Collapse
Affiliation(s)
- Ye Han
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Na Fu
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Wenjie Chen
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Jingjing Liang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Yanan Cui
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Ying Zhang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
49
|
De Wispelaere LA, Ouwehand S, Olsthoorn M, Govaert P, Smit LS, de Jonge RC, Lequin MH, Reiss IK, Dudink J. Electroencephalography and brain magnetic resonance imaging in asphyxia comparing cooled and non-cooled infants. Eur J Paediatr Neurol 2019; 23:181-190. [PMID: 30279083 DOI: 10.1016/j.ejpn.2018.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/29/2018] [Accepted: 09/04/2018] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The aim was to establish any differences in the predictive value of EEG and MRI for outcome in infants treated and not-treated with therapeutic hypothermia (HT) for perinatal asphyxia. We hypothesize that they are equally predictive and that combining both has the highest predictive value. STUDY DESIGN We retrospectively compared data of infants with hypoxic-ischemic encephalopathy (HIE) who received HT (n = 45) between September 2009 and December 2013 with those of infants with HIE born between January 2004 and August 2009, before HT was available (NT, n = 37). All received conventional and/or amplitude-integrated EEG during the first days and early MRI (day 4-5). Associations of EEG, MRI and severe neurodevelopmental outcome (death or Bayley's -2SD below mean), were tested with a multivariable logistic regression analysis, corrected for HT. RESULTS Forty-eight hours' EEG background pattern had a PPV of 92% and a NPV of 81% in HT, versus 100% and 58% in NT. MRI had a PPV of 71% and a NPV of 93% in HT, versus 82% and 75% in NT. The adjusted OR for adverse outcome was 0.013 (95% CI 0.002-0.154, p < 0.001) for EEG background normalization within 48 h and 32.19 (95% CI 4.84-214.25, p < 0.001) for abnormal MRI. CONCLUSION The predictive value of EEG and MRI is equal in cooled and non-cooled infants with HIE. Our data show a higher predictive value (death and severe outcome) for EEG compared to MRI. In HIE, persistent abnormal EEG background pattern until 48 h, combined with abnormal early MRI is strongly predictive for poor neurodevelopment.
Collapse
Affiliation(s)
- Leen Att De Wispelaere
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, the Netherlands; Department of Neonatology, University Hospital Antwerp, Wilrijkstraat 10, 2650 Edegem, Belgium.
| | - Sabine Ouwehand
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, the Netherlands.
| | - Marielle Olsthoorn
- Department of Radiology, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, the Netherlands.
| | - Paul Govaert
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, the Netherlands.
| | - Liesbeth S Smit
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, the Netherlands; Department of Neurology, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, the Netherlands.
| | - Rogier Cj de Jonge
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, the Netherlands; Department of Neonatology, VU University Medical Center, De Boelelaan 1105, 1081 HV, Amsterdam, the Netherlands.
| | - Maarten H Lequin
- Department of Radiology, Wilhelmina Children's Hospital, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| | - Irwin K Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, the Netherlands.
| | - Jeroen Dudink
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Wytemaweg 80, 3015 CN, Rotterdam, the Netherlands; Department of Pediatrics, Division of Neonatology, Wilhelmina Children's Hospital, University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| |
Collapse
|
50
|
Boylan GB, Kharoshankaya L, Mathieson SR. Diagnosis of seizures and encephalopathy using conventional EEG and amplitude integrated EEG. HANDBOOK OF CLINICAL NEUROLOGY 2019; 162:363-400. [PMID: 31324321 DOI: 10.1016/b978-0-444-64029-1.00018-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Seizures are more common in the neonatal period than at any other time of life, partly due to the relative hyperexcitability of the neonatal brain. Brain monitoring of sick neonates in the NICU using either conventional electroencephalography or amplitude integrated EEG is essential to accurately detect seizures. Treatment of seizures is important, as evidence increasingly indicates that seizures damage the brain in addition to that caused by the underlying etiology. Prompt treatment has been shown to reduce seizure burden with the potential to ameliorate seizure-mediated damage. Neonatal encephalopathy most commonly caused by a hypoxia-ischemia results in an alteration of mental status and problems such as seizures, hypotonia, apnea, and feeding difficulties. Confirmation of encephalopathy with EEG monitoring can act as an important adjunct to other investigations and the clinical examination, particularly when considering treatment strategies such as therapeutic hypothermia. Brain monitoring also provides useful early prognostic indicators to clinicians. Recent use of machine learning in algorithms to continuously monitor the neonatal EEG, detect seizures, and grade encephalopathy offers the exciting prospect of real-time decision support in the NICU in the very near future.
Collapse
Affiliation(s)
- Geraldine B Boylan
- Department of Paediatrics and Child Health, Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland.
| | - Liudmila Kharoshankaya
- Department of Paediatrics and Child Health, Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland
| | - Sean R Mathieson
- Department of Paediatrics and Child Health, Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|