You are on page 1of 14

Advances in Neonatal Acute Kidney

Injury
Michelle C. Starr, MD, MPH,a Jennifer R. Charlton, MD, MSc,b Ronnie Guillet, MD, PhD,c Kimberly Reidy, MD,d
Trent E. Tipple, MD,e Jennifer G. Jetton, MD,f Alison L. Kent, BMBS, FRACP, MD,c,g Carolyn L. Abitbol, MD,h
Namasivayam Ambalavanan, MD,i Maroun J. Mhanna, MD, MPH, MBA,k David J. Askenazi, MD, MSPH,j
David T. Selewski, MD, MS,l Matthew W. Harer, MD,m on behalf of the Neonatal Kidney Collaborative Board

In this state-of-the-art review, we highlight the major advances over the last 5 abstract
years in neonatal acute kidney injury (AKI). Large multicenter studies reveal
that neonatal AKI is common and independently associated with increased
morbidity and mortality. The natural course of neonatal AKI, along with the
risk factors, mitigation strategies, and the role of AKI on short- and long-term a
Division of Pediatric Nephrology, Department of
outcomes, is becoming clearer. Specific progress has been made in identifying Pediatrics, School of Medicine, Indiana University,
Indianapolis, Indiana; bDivision of Nephrology, Department
potential preventive strategies for AKI, such as the use of caffeine in premature of Pediatrics, University of Virginia, Charlottesville,
neonates, theophylline in neonates with hypoxic-ischemic encephalopathy, and Virginia; cDivision of Neonatology, Department of
Pediatrics, Golisano Children’s Hospital, University of
nephrotoxic medication monitoring programs. New evidence highlights the Rochester Medical Center, Rochester, New York; dDivision
importance of the kidney in “crosstalk” between other organs and how AKI of Pediatric Nephrology, Department of Pediatrics, Albert
Einstein College of Medicine, Bronx, New York; eSection of
likely plays a critical role in other organ development and injury, such as
Neonatal-Perinatal Medicine, Department of Pediatrics,
intraventricular hemorrhage and lung disease. New technology has resulted in College of Medicine, The University of Oklahoma, Oklahoma
advancement in prevention and improvements in the current management in City, Oklahoma; fDivision of Nephrology, Dialysis, and
Transplantation, Stead Family Department of Pediatrics,
neonates with severe AKI. With specific continuous renal replacement therapy University of Iowa Stead Family Children’s Hospital, Iowa
machines designed for neonates, this therapy is now available and is being City, Iowa; gCollege of Health and Medicine, The Australian
National University, Canberra, Australia Capitol Territory,
used with increasing frequency in NICUs. Moving forward, biomarkers, such as Australia; hDivision of Pediatric Nephrology, Department of
urinary neutrophil gelatinase–associated lipocalin, and other new technologies, Pediatrics, Miller School of Medicine, University of Miami
and Holtz Children’s Hospital, Miami, Florida; iDivisions of
such as monitoring of renal tissue oxygenation and nephron counting, will Neonatology and jNephrology, Department of Pediatrics,
likely play an increased role in identification of AKI and those most vulnerable University of Alabama at Birmingham, Birmingham,
Alabama; kDepartment of Pediatrics, Louisiana State
for chronic kidney disease. Future research needs to be focused on
University Shreveport, Shreveport, Louisiana; lDivision of
determining the optimal follow-up strategy for neonates with a history of AKI Nephrology, Department of Pediatrics, Medical University
to detect chronic kidney disease. of South Carolina, Charleston, South Carolina; and
m
Division of Neonatology, Department of Pediatrics, School
of Medicine and Public Health, University of
Wisconsin–Madison

Since the publication of the improved.5,6 In this state-of-the-art Drs Starr and Harer conceptualized and designed this
review, drafted the initial manuscript, and reviewed and
“Neonatal Acute Kidney Injury” review, we will review neonatal revised the manuscript; Drs Charlton and Selewski
conceptualized and designed this review, coordinated
review in 2015, our understanding of kidney physiology, provide an update and provided oversight, and reviewed and revised the
the epidemiology and impact of of neonatal AKI knowledge manuscript; Drs Guillet, Reidy, Tipple, Jetton, Kent,
Abitbol, Ambalavanan, Mhanna, and Askenazi provided
neonatal acute kidney injury (AKI) (definitions, prevalence, outcomes, substantial acquisition and assimilation of the data,
has exponentially increased.1 Single- and complications), discuss the drafted sections of the manuscript, and critically
revised the manuscript; and all authors approved the
center and multicenter work has current state of research, and final manuscript as submitted and agree to be
clearly revealed that AKI occurs appraise cutting edge data on accountable for all aspects of the work.

commonly in critically ill neonates therapeutics and devices that will DOI: https://doi.org/10.1542/peds.2021-051220

and adversely impacts outcomes.2–4 improve care in the coming decade. Accepted for publication Aug 13, 2021
In parallel to these advancements,
our ability to identify AKI early, NEONATAL KIDNEY PHYSIOLOGY AND
mitigate AKI, and provide renal IMPLICATIONS FOR AKI To cite: Starr MC, Charlton JR, Guillet R, et al.
Advances in Neonatal Acute Kidney Injury.
replacement therapy (RRT) with A basic understanding of kidney Pediatrics. 2021;148(5):e2021051220
devices designed for neonates has structure and function during

PEDIATRICS Volume 148, number 5, November 2021:e2021051220 STATE OF THE ART REVIEW
Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf
by Hospices Civils de Lyon user
development is essential to at birth and reaches adult levels by Since the publication of this
understand neonatal AKI and its 1 year of age.13 Poor urinary definition, there have been several
consequences.1 Nephrogenesis concentrating ability, particularly in observational studies highlighting
begins at 5 weeks’ gestation and neonates with high insensible losses potential areas for future refinement
continues until 34 to 36 weeks’ or critical illness, predisposes to account for chronological and
gestation.7 The nephron number is neonates to volume depletion and gestational age (GA). For example,
highly variable at birth, ranging subsequent prerenal azotemia. whereas some researchers advocate
from 200 000 to 2.7 million, and is Finally, neonatal kidneys appear to for excluding the SCr level from the
impacted by a multitude of factors, be particularly susceptible to first postnatal 48 hours when
including prematurity.8,9 ischemic injury to the renal tubules, calculating the baseline SCr level,
even after a mild and short-term others believe that this value is a
There are several core principles of insult. This is further complicated surrogate of the nephron number
neonatal physiology that uniquely when nephrotoxic medications, such and is clinically meaningful.2,3,17–19
impact the diagnosis and as gentamicin and other Researchers have also pointed out
management of neonatal AKI. First, aminoglycosides, are commonly that deviations in the GA-
both renal blood flow and perfusion prescribed to critically ill neonates appropriate SCr trajectory (which
pressure increase over the first and result in tubular injury. steadily drops from birth in healthy
weeks of life in neonates. The term neonates) could signify AKI,
proportion of cardiac output DEFINITIONS OF AKI but this is not captured in the
delivered to the kidneys increases current KDIGO definition.20
The neonatal modified Kidney
from 5% during fetal life to 20% by
Disease: Improving Global Outcomes The current neonatal modified
2 years.10 Much of this increased
(KDIGO) definition is the most KDIGO AKI definition incorporates
blood flow occurs after birth, with
commonly used definition used in UOP. Despite studies in older
renal blood flow doubling in the
clinical practice and most populations revealing that UOP is
first 2 postnatal weeks. After birth,
epidemiological studies (Table 1). critically important to properly
the distribution of blood flow
This empirical definition stages AKI identify AKI, few studies in neonatal
transitions from deeper, more
severity on the basis of a rise in AKI have included UOP. One study
mature glomeruli to superficial,
serum creatinine (SCr) levels from a using diaper weights every 3 hours
cortical glomeruli.11 This change in
blood flow can be altered by
previous trough and/or a decrease in found that UOP <1.5 mL/kg per
urine output (UOP).14,15 The National hour was associated with increased
medications (such as indomethacin),
Institutes of Health–sponsored mortality.21 Furthermore, they
perinatal asphyxia, and maternal
Neonatal AKI Workshop in 2013 found that lower thresholds of UOP
hemorrhage, which all predispose
recommended that researchers and (<1 mL/kg per hour) were
neonates to AKI. Second, congruent
clinicians use the neonatal modified associated with an even higher
with the increased blood flow, the
KDIGO definition to define AKI but mortality rate.21 Few studies,
glomerular filtration rate (GFR)
emphasized that this definition should including the Assessment of
increases dramatically after birth
be a starting point for an iterative Worldwide Acute Kidney Injury
and reaches adult levels by 2 years
process, which is based on clinically Epidemiology in Neonates
of age.12 The GFR is low in infants,
meaningful and long-term (AWAKEN) study, included UOP
both in absolute value and when
outcomes.16 measurement in the assessment of
corrected for body surface area
(milliliters per minute per 1.73
TABLE 1 Neonatal AKI KDIGO Classification
meters2). For example, premature
infants born at 26 weeks have a GFR Stage SCr UOP
as low as 0.7 mL/minute per kg on 0 No change in SCr level or rise <0.3 mg/dL $0.5 mL/kg per h
day 1 of age, which improves only 1 Increase in SCr level of $0.3 mg/dL within 48 h <0.5 mL/kg per h for 6–12 h
slightly during the first several or rise in SCr level $1.5–1.9 times the
reference SCr levela within 7 d
weeks of life.12 In neonates with a 2 Rise in SCr level $2–2.9 times the reference SCr <0.5 mL/kg per h for $12 h
physiologically low GFR, additional levela within 7 d
stressors, such as sepsis, hypoxia, 3 SCr level $3 times the reference SCr levela or <0.3 mL/kg per h for $24 h or
hypotension, or other clinical SCr level >2.5 mg/dLb or receipt of RRT anuria for $12 h
a
conditions common in prematurity, Differences between the neonatal AKI definition and KDIGO definition: reference SCr level defined as the lowest pre-
vious SCr value.
may increase the risk AKI. Third, b
Differences between the neonatal AKI definition and KDIGO definition: SCr value of 2.5 mg/dL represents <10 mL/
urinary concentrating ability is low min per 1.73m2.

2 STARR et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf
by Hospices Civils de Lyon user
AKI, thus limiting the available data notable studies evaluating the occurred in 45% of neonates <29
on which to base thresholds for epidemiology and impact of weeks’ GA and in 14% of neonates 29
diagnosis and determination of neonatal AKI in the last 5 years to 36 weeks’ GA. In the AWAKEN
severity of AKI. In future studies, (Table 3). Much of this increased study, the potentially modifiable risk
researchers should carefully knowledge stems from the AWAKEN factors for early AKI in ELGANs were
measure UOP to determine the study, which enrolled neonates at mainly medication exposures.25 Given
impact of UOP on AKI diagnosis in risk for AKI (determined by >48 immature tubular function and
neonates. hours of intravenous fluids). In this resulting poor urinary concentration
cohort, the risk of AKI occurred in a ability, along with the increased
Given the interest and focus on AKI bimodal pattern, with extremely low insensible losses common in preterm
diagnostic thresholds, it is likely that gestational age neonates (ELGANs) neonates, volume depletion leading to
a refined definition of neonatal AKI (<28 weeks) and term infants at the prerenal azotemia is a common factor
will emerge. Two recent greatest risk.4 The AWAKEN study predisposing this population to AKI.
publications, including an analysis of identified a clear variation in SCr
the AWAKEN study, suggest an monitoring practices across centers, Patent ductus arteriosus (PDA) is an
alternative definition for neonatal with less than half of centers important clinical issue for preterm
AKI.22,23 In this study, the authors checking $5 SCr levels during infants and is associated with a
proposed different cutoffs for the hospital admission. Not surprisingly, higher risk of AKI.26,27 PDA
first postnatal week by GA, the rates of AKI by center were represents a clinical challenge
compared with the subsequent directly correlated with the average because AKI may result if it is left
weeks, and provided cutoffs by GA number of SCr samples ascertained
group.23 For example, in infants untreated; however, classic PDA
per subject.4 This practice variation
#29 weeks’ GA, a rise in the SCr therapies may also be associated
is particularly notable in the context
level of 0.6 mg/dL confers the with AKI. Although nonsteroidal
of the recent Baby Nephrotoxic
highest prediction of mortality, antiinflammatory drug (NSAID)
Injury Negated by Just-in-Time
whereas in infants >29 weeks’ GA, a treatment of PDA added an
Action (NINJA) publication, which
rise of 0.3 mg/dL is of highest additional risk for mild AKI, severe
revealed that by monitoring SCr
mortality prediction.23 Furthermore, more frequently in neonates with
AKI was less likely when NSAID
we anticipate that novel approaches high nephrotoxic medication treatment was effective.26
of using urine biomarkers, SCr exposure, there was an increased Term or Near-Term Critically Ill
thresholds, UOP thresholds, and awareness of the risk for AKI that, Neonates
fluid balance metrics will be used to in turn, resulted in a lower rate and
enhance the current neonatal KDIGO In the AWAKEN study, the incidence
duration of AKI.17 These data
definition. In the interim, we of AKI in neonates born at $36
suggest that critically ill neonates
recommend that the neonatal may benefit from protocolized SCr weeks and admitted to a NICU was
modified KDIGO definition be used monitoring during high-risk events. 37%.4 The etiology of AKI in term
as the standard until newer neonates is often multifactorial and
definitions are widely validated in Preterm Neonates includes risk factors related to their
large multisite trials and correlated The risk of AKI increases markedly illness and management
with long-term outcomes. with decreasing GA.2,3 In cohorts of (Table 2).25,28 Multiorgan
very low birth weight (VLBW) dysfunction is common and occurs
EPIDEMIOLOGY, RISK FACTORS, AND neonates, the incidence of AKI is in up to 70% of neonates with
ASSOCIATED FINDINGS WITH AKI reported between 18% and 40%.3,24 AKI.29–31 Some of the major risk
AKI is common in critically ill In ELGANs enrolled in the Preterm factors for AKI include hypoxic-
neonates. We present a summary of Erythropoietin Neuroprotection ischemic encephalopathy (HIE),
the risk factors associated with Trial, 38% had at least 1 episode of cardiac disease, surgery, and
neonatal AKI (Table 2) and the most AKI.2 In the AWAKEN study, AKI nephrotoxic medications.

TABLE 2 Epidemiology, Risk Factors and Associated Findings with Neonatal AKI
Prenatal Perinatal Postnatal
Factors that increase risk of a preterm or LBW Exposure to nephrotoxic medications (ACE Prematurity, LBW, CHD, inborn errors of
neonate, placental insufficiency inhibitors, NSAIDs), delivery complications metabolism, sepsis, nephrotoxin exposure,
resulting in hypoxia and/or asphyxia, HIE PDA, extracorporeal therapies
ACE, angiotensin-converting enzyme; LBW, low birth weight.

PEDIATRICS Volume 148, number 5, November 2021 3

Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf


by Hospices Civils de Lyon user
TABLE 3 Summary of Neonatal AKI Studies Published Between 2015 and 2020
Study Design and Population
Author (Year), N Details Main Findings
4
Jetton et al (2017), Neonates requiring >48 h of AKI occurred in 45% of the infants at #28 wk GA and in 14% of infants at 29–36
N 5 2162 intravenous fluids in the NICU wk GA.
Charlton et al25,28 Secondary analysis of infants Antimicrobial agents, methylxanthines, diuretics, NSAIDs, hypertensive disorders of
(2019), N 5 2162 from the AWAKEN study pregnancy, and hypoglycemia were associated with lower odds of early AKI. In
infants at 29–25 wk GA, outborn status, saline bolus during resuscitation, and
more frequent SCr monitoring were associated with higher odds of early AKI.
Askenazi et al2 Prospective study of ELGANs in Thirty-eight percent had at least 1 episode of stage 1 or higher AKI, and 18.2%
(2000), N 5 923 the PENUT trial had 1 episode of stage 2 or higher AKI.
Selewski et al51 Retrospective analysis of infants Median peak fluid balance was 1.0% (IQR: 0.5 to 4.6) over the first postnatal
(2019), N 5 645 at $36 wk GA from the week and occurred on postnatal d 3 (IQR: 1 to 5). Mechanical ventilation on
AWAKEN study postnatal d 7 was associated independently with the following measures of
fluid balance over the first postnatal week: peak fluid balance (aOR: 1.12; 95%
CI: 1.08 to 1.17), lowest fluid balance in first postnatal week (aOR: 1.14; 95% CI:
1.07 to 1.22), fluid balance on postnatal d 7 (aOR: 1.12; 95% CI: 1.07 to 1.17),
and negative fluid balance at postnatal d 7 (aOR: 0.3; 95% CI: 0.16 to 0.67).
Selewski et al53 Retrospective analysis of infants Median peak fluid balance was 0% (IQR: 2.9 to 2) and occurred on postnatal d 2 (IQR:
(2020), N 5 1007 at <36 wk GA from the 1 to 5). Mechanical ventilation on postnatal d 7 was associated independently with
AWAKEN study the following measures of fluid balance over the first postnatal week: peak fluid
balance (aOR: 1.14; 95% CI: 1.10 to 1.19), lowest fluid balance (aOR: 1.12; 95% CI: 1.07
to 1.16), fluid balance on postnatal d 7 (aOR: 1.10; 95% CI: 1.06 to 1.13), and negative
fluid balance at postnatal d 7 protected against the need for mechanical ventilation
at postnatal d 7 (aOR: 0.21; 95% CI: 0.12 to 0.35).
Nour et al120 (2020), Retrospective analysis in No difference in AKI between the those receiving selective head cooling and those
N 5 30 neonates with HIE who not. SCr levels and UOP were significantly improved on d 4 and d 10 samples
underwent cooling via compared with baseline samples in both groups regardless of cooling. There
selective head cooling was a difference in NGAL levels, but not CysC levels, on d 4 and 10.
Bellos et al90 (2019), Meta-analysis of effectiveness of Incidence of AKI significantly lower in neonates receiving theophylline (OR: 0.24;
N 5 458 theophylline administration in 95% CI: 0.16 to 0.36), whereas mortality rates were similar between the 2
neonates with perinatal groups (OR: 0.86; 95% CI: 0.46 to 1.62). Theophylline administration was
asphyxia associated with significantly decreased SCr levels (MD: 0.57 mg/dL; 95% CI:
0.68 to 0.46) in the third day of life.
Harer et al89 (2018), Retrospective analysis of AKI occurred less frequently in neonates who received caffeine in the first week
N 5 675 premature infants at <33 wk of life than in those who were not treated with caffeine (11.2% vs 31.6%; P <
GA from the AWAKEN study .01). Neonates who received caffeine had more AKI risk factors, including lower
average GA, lower birth wt, and high severity of illness scores, and were still
less likely to develop stage 2 or 3 AKI. The No. needed to treat with caffeine to
prevent 1 episode of AKI was 4.3.
Stoops et al17 Prospective quality improvement Reduced high nephrotoxic medication exposures from 16.4 to 9.6 per 1000
(2019) effort to reduce nephrotoxic patient-days (P 5 .03), reduced nephrotoxic medication–associated AKI from
medication–associated AKI in 30.9% to 11.0% (P < .001), and reduced AKI severity from 9.1 to 2.9 per 100
the NICU susceptible patient-days (P < .001) prevented 100 AKI episodes during the 18-
mo sustainability era.
Starr et al103 (2019), Retrospective analysis of Infants born between 29 and 32 wk GA with AKI had fourfold higher odds of
N 5 546 premature infants at <32 wk moderate or severe BPD or death after controlling for multiple factors (aOR:
GA from the AWAKEN study 4.21; 95% CI: 2.07 to 8.61).
aOR, adjusted odds ratio; BPD, bronchopulmonary dysplasia; IQR, interquartile range; MD, mean difference; PENUT, Preterm Erythropoietin Neuroprotection Trial.

HIE with stage III HIE having AKI, surgery for congenital heart disease
There is a general agreement that compared with 7.4% of those with (CHD).34–36 One single-center
the presence of AKI in the setting of stage II HIE.32 retrospective study of neonates with
HIE is associated with poor single ventricle physiology
Cardiac Disease and Extracorporeal undergoing stage 1 Norwood
outcomes (increased mortality, poor
neurodevelopmental outcomes,
Membrane Oxygenation palliation found that 21% developed
longer hospital stay, and longer Infants who require cardiac surgery AKI.36 A large Danish registry study
duration of mechanical and those who need extracorporeal revealed that 33% of neonates had
ventilation).30,32,33 In addition, there membrane oxygenation (ECMO) are AKI within 5 days of surgery.35 In a
is a correlation between severity of at high risk for AKI. AKI occurs in multicenter retrospective cohort
HIE and AKI, with 70% of those 30% to 50% of patients undergoing study of 832 pediatric patients on

4 STARR et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf
by Hospices Civils de Lyon user
ECMO, 74% had AKI.34 AKI was FLUID BALANCE standardized weight measurement
present at initiation of ECMO in the The development of fluid overload is protocols to properly perform the
majority of cases and was associated an independent predictor of adverse weight-based technique.52,54
with a longer ECMO duration and outcomes across pediatric critical
increased mortality.34 The risk of care populations.40–49 The Fluid balance in the early postnatal
AKI in those on ECMO varies by development of fluid overload is period can be challenging to
underlying diagnosis; those with often multifactorial, resulting from interpret in the setting of normal
congenital diaphragmatic hernia AKI, iatrogenic fluid administration, postnatal diuresis and expected
were more likely to require RRT.37 capillary leak from systemic negative fluid balance. Although an
inflammation, and aberrant average weight loss of 7% from
Surgery homeostatic mechanisms.41 birth weight is described in term
The incidence of AKI is high in Additionally, fluid overload can neonates, the normal fluid balance
noncardiac surgery; 34% of neonates make the diagnosis of AKI more for neonates of other GAs is less
challenging because SCr is diluted in clearly defined, particularly in
undergoing abdominal and thoracic
extremely preterm neonates with
surgery have an episode of AKI.38 the setting of a positive fluid
balance, resulting in a potential excessive skin permeability.55 Early
Infants with AKI after surgery were
underdiagnosis of AKI.41 Precise positive postnatal fluid balance is
more likely to have VLBW. They were
definitions are essential to associated with adverse short-
also more likely to have sepsis, a
understanding the epidemiology and (death, mechanical ventilation on
longer duration of mechanical
impact of excessive fluid day 7) and long-term outcomes
ventilation, an operative time >120
accumulation on outcomes in (bronchopulmonary dysplasia) in
minutes, necrotizing enterocolitis
neonates (Table 4).50 The 2 most neonates.51,53,56–59 There remains a
(NEC), and a higher risk of
common methods used to calculate paucity of data defining the
mortality.38 Among infants with
fluid balance are the cumulative pathologic state of fluid overload in
surgically managed NEC, almost 60%
fluid balance and weight-based critically ill neonates, which in older
had severe AKI (stage 2 or 3).39
methods. The weight-based method children, is commonly defined as a
Nephrotoxic Medications to describe fluid balance is used to cumulative positive fluid balance of
calculate the degree of fluid $10% to 20%.48,60 Multiple critical
Many neonates are exposed to gaps exist in our understanding of
overload on the basis of a change in
nephrotoxic medications in the the causes and impact of abnormal
weight from a baseline weight (birth
NICU, which can contribute to AKI. fluid balance in neonates. These
weight or determined dry weight)51:
Although there are many gaps include interpreting fluid
daily fluid balance 5 change in daily
nephrotoxic medications, these balance in neonates, especially those
weight day to day; cumulative fluid
primarily include antimicrobial
balance 5 daily weight baseline who have spent a considerable time
agents (eg, acyclovir, amphotericin
weight; percentage fluid overload44 5 in the NICU. Research is greatly
B, aminoglycosides, needed to understand the optimal
([daily weight baseline weight]/
vancomycin).25,28 It was recently
baseline weight)  100. threshold to define appropriate fluid
reported in Baby NINJA, a single- balance and to understand the
center quality improvement Although each method has been detrimental effects of neonatal fluid
program focused on reducing used in older children, the weight- overload on extrarenal organ
nephrotoxic medication–associated based methods represent the systems (eg, oxygenation index and
AKI, that attention to high-risk standard in neonates because fluid cardiac dysfunction due to excessive
neonates, including daily SCr balances have been shown to be fluid and reduced contractility).
monitoring, reduces nephrotoxic inaccurate.51–53 It is critical to use Understanding the role of fluid
medication exposures (P 5 .03),
nephrotoxic medication–associated TABLE 4 Definitions of Excessive Fluid Accumulation in Neonates
AKI (P < .001), and AKI duration (P
Term Definition
< .001).17 This suggests that
identification and monitoring of Daily fluid balance Daily difference between input and output or change in wt over a 24-
h period
high-risk neonates, with a thoughtful
Cumulative fluid balance Change in fluid balance over a given duration
consideration of nephrotoxic Fluid overload Cumulative fluid balance expressed as a percentage of body wt; used
medications, may minimize AKI and to refer to the pathologic state of excessive fluid accumulation
its consequences in critically ill associated with the development of sequelae attributable to fluid
accumulation and adverse outcomes
neonates.

PEDIATRICS Volume 148, number 5, November 2021 5

Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf


by Hospices Civils de Lyon user
balance in various neonatal filtered by glomeruli and reabsorbed predicting AKI in critically ill
populations (premature versus term in the proximal tubule. Although neonates.71 This combination has
neonates) at critical time points both serum and urinary CysC have been evaluated in pediatric and
(perinatal versus postnatal versus been assessed as early markers of adult populations and has been
postsurgical) in the context of AKI, a rise in the serum CysC level shown to perform well in predicting
underlying disease process (NEC, reflects a change in kidney function, subsequent severe AKI.63 Both NGAL
lung disease, and sepsis) is a critical whereas an elevated urinary CysC and TIMP-2 and IGFBP-7 are
knowledge gap. Answers to these level is considered reflective of currently used off label in multiple
questions will drive therapeutic tubular injury.12,64–68 A recent pediatric and adult ICUs to evaluate
interventions designed to prevent systematic review of neonates for AKI, and there are several
and mitigate harm from fluids. across all GA groups suggested that ongoing studies assessing their
serum CysC may be superior to SCr utility in pediatric AKI.72 Kidney
NEW ADVANCES IN AKI RESEARCH for assessing the GFR.65 injury molecule 1 is a
Furthermore, an elevated urinary transmembrane protein that is
Biomarkers of AKI CysC level has been shown to have upregulated in kidney injury, and
Proteins and metabolites are 2 an area under the curve of 0.85 elevated levels have been shown to
examples of biomarkers that can be (95% confidence interval [CI]: 0.81 predict AKI.73 By using SCr as the
measured consistently and correlate to 0.88) in predicting a rise in the gold standard, the areas under the
with the disease occurrence or SCr level 24 to 96 hours later in curve of several of these biomarkers
progression.61 Given the diversity of neonates post surgery or after (in particular, NGAL and TIMP-2
GA and etiologies of AKI in perinatal asphyxia.67 and IGFBP-7) are highly predictive
neonates, 1 biomarker does not for AKI.73 It is imperative that we
appear to reliably predict AKI. There are several other biomarkers begin to incorporate novel
However, more reference ranges are that may help identify injury and biomarkers into future definitions
becoming available for novel urinary functional changes before of neonatal AKI and into clinical
biomarkers by GA and postnatal permanent damage in neonates with care.
age.62 SCr represents the current AKI. Neutrophil
gelatinase–associated lipocalin Nephron Number
standard for diagnosing neonatal
AKI. However, it is critical to (NGAL) is a protein bound to In humans, there is wide variability
understand the shortcomings of SCr neutrophil granules, filtered by the in the nephron number present,
and develop novel biomarkers that glomerulus, and reabsorbed by the even in the neonatal period because
fill these gaps. A rise in the SCr level proximal tubules.69 NGAL is highly the completion of nephrogenesis
indicates a loss of kidney function, sensitive (87%–93%) and specific occurs at 26 to 34 weeks.8,9,74 The
reflecting injury that occurred up to (87%–93%) for AKI in neonates mechanisms (in utero and ex utero)
48 to 72 hours before.63 As a result, with perinatal asphyxia.70 The that influence nephrogenesis, and
biomarker studies, such as the ones combination of G1 cell cycle arrest ultimately nephron endowment, and
below, have been focused on biomarkers urinary tissue inhibitor the processes regulating nephron
identifying injury and functional of metalloproteinase 2 (TIMP-2) and loss are poorly understood.7
changes before permanent damage insulinlike growth factor binding Preclinical advancements have been
(Table 5). protein 7 (IGFBP-7) is promising for made in measuring the whole-
detecting early AKI. It has been kidney functional nephron number
Cystatin C (CysC) is a cysteine shown to have a sensitivity of 89% in vivo with a novel contrast agent.
protease inhibitor that is freely and a specificity of 51% for Cationic ferritin-enhanced MRI has

TABLE 5 Biomarkers in Use or Under Study to Assist With AKI Prediction and Diagnosis
Biomarker Location of Injury Notes
SCr Reflects kidney function, not injury; metabolic product of Delayed marker (48–72 h) of kidney function
skeletal muscle creatine
CysC Reflects kidney function, not injury (serum); proximal Increase in the serum CysC level is thought to reflect a change
tubule injury (urine) in GFR or kidney function, whereas an elevated urinary CysC
level is considered reflective of tubular injury
NGAL Distal tubule and collecting duct Highly sensitive for ischemia and nephrotoxins
Kidney injury molecule 1 Proximal tubule injury Regulates apoptosis, promotes epithelial regeneration
TIMP-2 and IGFBP-7 Tubule cells Limits proliferation of damaged tubule cells; marker of early AKI

6 STARR et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf
by Hospices Civils de Lyon user
been used to measure the (interventions required at delivery), high-risk neonatal cohorts to
glomerular number and size in nephrotoxic medications exposure, prevent AKI. There have been
health and disease.75,76 The radial and postnatal events. Physical multiple therapeutics evaluated for
glomerular count, a surrogate examination should include an AKI prevention in neonates (without
marker of glomerulogenesis, assessment of volume status, which positive results), including
suggests that the nephron number is should also include weight, daily erythropoietin, therapeutic
decreased in both premature fluid balance, and cumulative fluid hypothermia, remote ischemic
neonates and in those with AKI.77 balance. Fluid balance assessment is preconditioning, and
Further work is necessary to essential because volume depletion corticosteroids.86–88
translate this technique to humans, is a common cause of AKI and Methylxanthines have been
including neonates, to further volume overload is a common evaluated in multiple neonatal
understand those at risk for future complication of AKI. Positive fluid populations and have had promise
chronic kidney disease (CKD).78 balance still may mean poor renal as a preventive treatment of AKI in
perfusion if neonates have ongoing high-risk populations.89,90
Tissue Oxygenation third spacing due to capillary leak.
Noninvasive continuous monitoring Maintaining euvolemia is a After diagnosis of AKI, careful
of renal oxygen saturation with challenging but essential management of fluid balance and
near-infrared spectroscopy (NIRS) is management strategy in infants both medications is essential to
a new diagnostic tool that may lead to prevent AKI and to mitigate preventing the development of
to earlier diagnosis of AKI.79 Renal severe volume overload and complications. Strict documentation
tissue oxygenation (RrSO2) complications. Focused laboratory of all fluid input and output, along
monitoring is a surrogate for local evaluation should be performed, with daily weights, is essential to
tissue oxygen use. In neonates with including measurement of serum optimizing fluid balance.
CHD, NIRS monitoring of the kidney electrolytes and serum urea Nephrotoxic medications should be
postoperatively can predict AKI.80 In nitrogen, as well as of SCr and/or assessed daily and reduced or
premature neonates, those who CysC for GFR assessment.65 At this eliminated whenever possible.17
subsequently develop AKI have time, there is not a definitive role Cumulative fluid balance should be
lower RrSO2 in the first postnatal for urine biomarker assessment in carefully monitored to assess overall
day or week.81,82 In postoperative all neonates, but growing data volume status. Infants with volume
cardiac patients, NIRS detected a suggest that it may be useful in depletion may require additional
decline in RrSO2 before AKI was certain clinical settings to predict fluid in the form of either enteral
defined by SCr or UOP.83 In those AKI.71 Fractional excretion of feeding, intravenous boluses, or
undergoing therapeutic hypothermia sodium may be helpful in some drips. In infants with volume
for HIE, neonates with AKI had infants in differentiating volume overload, diuretics can be trialed to
higher RrSO2 values, likely pointing depletion from intrinsic causes of maintain UOP.91 Response to
to a different cause or type of injury, AKI but can be challenging to furosemide (furosemide stress test)
than preterm infants or infants with interpret in premature infants has been used as a functional
CHD.84 Further work is needed to because of tubular immaturity.12 We biomarker for predicting severe
establish normative RrSO2 values in recommend that an ultrasound be AKI.92 Although furosemide has not
neonatal populations and treatment obtained to evaluate for evidence of been evaluated in all neonatal
guidelines incorporating RrSO2 obstruction as well as congenital populations, term infants with CHD
with a lower response to furosemide
values. abnormalities of the kidney. An
(median UOP at 2 hours after
ultrasound can also determine
furosemide treatment 1.2 vs 3.4 mL/
EVALUATION OF NEONATAL AKI kidney size. However, more studies
kg per hour; P 5 .01) have an
Evaluating a neonate who develops are needed to know if kidney size is
increased risk for persistent AKI.
AKI requires a systematic approach, helpful in understanding clinically
Although furosemide responsiveness
which includes consideration of kidney-related meaningful
is a potential functional marker of
common factors contributing to AKI. outcomes.85
kidney status, more studies in
A detailed history should be neonates are needed to standardize
obtained to assess for risk factors MANAGEMENT OF NEONATAL AKI the dose and definitions used.
for AKI, including birth weight and Although the search for treatments
GA, antenatal events (including or interventions for established Theophylline and its related salt,
prenatal ultrasounds), pregnancy neonatal AKI has remained elusive, aminophylline, have had success in
complications, birth history medications have been evaluated in increasing UOP and may prevent

PEDIATRICS Volume 148, number 5, November 2021 7

Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf


by Hospices Civils de Lyon user
AKI in infants with HIE. uremia refractory to medical both the availability and acceptance
Theophylline is an adenosine management. The 2 most common of therapies by decreasing the
receptor antagonist that prevents modalities for RRT in neonates are extracorporeal volume and improving
AKI by inhibiting adenosine- peritoneal dialysis (PD) and fluid removal precision.97
induced renal vasoconstriction. A continuous renal replacement therapy
recent meta-analysis of 7 (CRRT). CRRT can be added to the In recent years, industry has made
randomized controlled trials (458 extracorporeal circuit in infants significant innovations in neonatal
neonates with asphyxia not receiving ECMO therapy. Between PD RRT by developing neonatal-specific
receiving therapeutic and CRRT, the choice often depends CRRT machines and repurposing
hypothermia) found that on the available resources, center machines for neonatal use.98 The
theophylline administration was experience, and patient Cardio-Renal Pediatric Dialysis
associated with a significantly characteristics.95 These therapies are Emergency Machine (Medtronic,
lower incidence of AKI (odds ratio complementary in that some Minneapolis, MN) was approved for
[OR]: 0.24; 95% CI: 0.16 to 0.36).90 neonates or situations will have a use in children between 2.5 and
On the basis of this evidence, a higher chance of success with PD, 8 kg the United States in 2020 and
single dose of theophylline within whereas others will benefit from has been used in multiple countries
the first 6 postnatal hours in CRRT. PD remains a common first outside the United States. This
newborns with HIE is endorsed in choice for RRT in most institutions machine was developed specifically
the 2012 KDIGO guidelines to because it does not require vascular for neonates and small children.5 In
prevent AKI.15 Aminophylline has access, is more available, and is often contrast, Aquadex (Aquadex
also had promise as a rescue technically easier in the smallest FlexFlow; CHF Solutions, Eden
therapy in neonates with AKI patients.95 PD can be performed by Prairie, MN) is an ultrafiltration
treated with therapeutic using a temporary catheter if the RRT device designed for adults but with
hypothermia.93 requirement is thought to be short- an extracorporeal vol of 33 mL,
term. Depending on the catheter used, making it ideal to adapt to safely
Caffeine is also an adenosine PD can be successful in neonates as provide CRRT to infants.5 Successful
receptor antagonist that has been small as 830 g.96 CRRT using Aquadex has been
evaluated for renoprotective effects reported in infants as small as 1.4
in preterm cohorts. Two studies CRRT may be preferred in kg.5 These machines and others,
revealed that AKI occurred less hemodynamically unstable infants, such as the Newcastle Infant
frequently in VLBW infants and those with a history of abdominal Dialysis and Ultrafiltration System
preterm infants <33 weeks’ GA who surgery or NEC (which makes PD (Newcastle National Health Service
received caffeine within the first technically challenging), or those in Foundation Trust, Newcastle, United
postnatal week. In a retrospective whom tight control of volume status Kingdom), have begun to
study of 140 VLBW neonates,94 AKI is necessary. In the past, the sole revolutionize the field of CRRT for
occurred less frequently in those availability of CRRT equipment neonates by lowering the associated
who received caffeine (17.8% vs designed for adults and larger risks and will change the
43.6%; P 5 .002). In a secondary children presented a challenge to conversation about when and in
analysis of the AWAKEN study, AKI perform CRRT in neonates. This whom to initiate CRRT.6,99
occurred less frequently in neonates necessitated the use for larger
<33 weeks GA who received catheters, tubing, and filters, resulting COMPLICATIONS OF AKI
caffeine in the first postnatal week in high extracorporeal volumes and
(11.2% vs 31.6%, P < .01).89 On greater hemodynamic instability, Cross Talk Between AKI and Other
the basis of these data, the number often requiring either blood Organs
that needed to be exposed to transfusions or blood priming with AKI has been shown to adversely
caffeine to prevent 1 episode of each circuit change. Recent advances impact other organs.100 Initially
AKI was 4.3.89 have made CRRT increasingly thought to be only an association,
accessible and successful for studies suggest a causal relationship
RRT neonates. Introduction of smaller in which AKI appears to drive other
RRT remains the primary therapy filters, such as the HF-20 (total organ dysfunction and vice-versa,
for the complications of severe AKI. extracorporeal vol of 60 mL), which referred to as “crosstalk.”101
The indications for RRT in neonates was recently approved by the US Experimental models describe a
include acidosis, fluid overload, Food and Drug Administration for use lung-focused inflammatory process,
electrolyte abnormalities, and in the United States, has improved driven in part by cytokines such as

8 STARR et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf
by Hospices Civils de Lyon user
interleukins after AKI, which are neonates.86,114,115 The lack of for neonates at risk for short- and
deleterious to the lungs.102 Both appropriately powered studies, long-term kidney-related dis-
preterm and term infants with AKI consensus definitions for AKI and ease.98,119
have worse lung outcomes than CKD, and a consistent follow-up
their peers without AKI, including period are barriers to clearly
longer durations of mechanical defining the relationship between
ventilation and higher rates of neonatal AKI and subsequent CKD. ABBREVIATIONS
bronchopulmonary Although the Chronic Kidney
AKI: acute kidney injury
dysplasia.103,104 Neonatal AKI has Disease in Children study manages
AWAKEN: Assessment of
been shown to be an independent children with CKD and includes
Worldwide Acute
risk factor for neurologic information on birth weight, it does
Kidney Injury
complications, such as not include detailed data on Epidemiology in
intraventricular hemorrhage, poor neonatal course and AKI. Large Neonates
long-term neurocognitive multicenter long-term follow-up CHD: congenital heart disease
outcomes, and cardiovascular studies of neonates after AKI are CI: confidence interval
disease (hypertension).105,106 needed to completely understand CKD: chronic kidney disease
the future risk of CKD. CRRT: continuous renal
Risk of CKD After AKI
replacement therapy
The risk of developing CKD or end- CONCLUSIONS CysC: cystatin C
stage kidney disease after AKI is
Dramatic advances in the diagnosis ECMO: extracorporeal membrane
well detailed in adults.107 The
and epidemiology of neonatal AKI oxygenation
evidence of progression from AKI to
and our ability to care for neonates ELGAN: extremely low
CKD is less established in children
with kidney disease have occurred gestational age neonate
with AKI. In a systematic review of
in the last decade.116 New GA: gestational age
346 children (mean follow-up 6.5 GFR: glomerular filtration rate
technologies and therapies designed
years), the incidence of an abnormal HIE: hypoxic-ischemic
GFR <90 mL/minute per 1.73 m2 to prevent and treat neonatal AKI
augment these findings. Future encephalopathy
was 6.3% (95% CI: 5.1 to 7.5).108 IGFBP-7: insulinlike growth
The mechanisms for progression to work, including interventional trials
of therapeutics to treat AKI factor binding protein 7
CKD are incompletely understood KDIGO: Kidney Disease
but likely are secondary to (methylxanthines, RRT with novel
devices), prospective long-term Improving Global
maladaptive repair, ongoing Outcomes
inflammation, and disordered follow-up studies to understand risk
factors for CKD development, and NEC: necrotizing enterocolitis
regeneration.109,110 Histologic NGAL: neutrophil
findings of preterm neonates reveal improved definitions of fluid
gelatinase–associated
abnormal glomeruli likely to develop overload, is needed. Additionally,
lipocalin
sclerosis later, which could be the continued integration of biomarkers
NINJA: Nephrotoxic Injury
explanation for later CKD in those into routine clinical use, more
Negated by Just-in-Time
with AKI.111 These changes may be widespread availability and use of
Action
superimposed on a decreased neonatal-specific extracorporeal
NIRS: near-infrared spectroscopy
nephron number and reduction in devices for kidney support therapy,
NSAID: nonsteroidal
future development of nephrons due and standardization of monitoring
antiinflammatory drug
to prematurity.74 and follow-up of neonates with AKI
OR: odds ratio
will continue to advance the field of
PD: peritoneal dialysis
The evidence for progression from neonatal AKI.117,118 Ongoing
PDA: patent ductus arteriosus
AKI to CKD in neonates is less clear. collaboration between
RrSO2: Renal tissue oxygenation
Several studies have identified neonatologists, pediatricians, and
RRT: renal replacement therapy
evidence of kidney abnormalities in nephrologists (including the SCr: serum creatinine
preterm infants with a history of Neonatal Kidney Collaborative; TIMP-2: tissue inhibitor of
AKI.112,113 In contrast, other studies www.babykidney.org) will help metalloproteinase 2
have failed to identify differences in drive these research initiatives, UOP: urine output
CKD or GFR in follow-up of preterm mentor young faculty, educate clini- VLBW: very low birth weight
infants who had AKI as cians, inform families, and advocate

PEDIATRICS Volume 148, number 5, November 2021 9

Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf


by Hospices Civils de Lyon user
Address correspondence to Michelle C. Starr, MD, MPH, School of Medicine, Indiana University and Riley Hospital for Children, 699 Riley Hospital Dr, RR230, Indianapolis, IN 46202.
E-mail: mcstarr@iu.edu
PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online, 1098-4275).
Copyright © 2021 by the American Academy of Pediatrics
FINANCIAL DISCLOSURE: The authors have indicated they have no financial relationships relevant to this article to disclose.

FUNDING: No external funding.

POTENTIAL CONFLICT OF INTEREST: For full disclosure, we provide here an additional list of other authors’ commitments and funding sources that are not directly related to this
study: Dr Askenazi is a consultant for Baxter, Nuwellis, Medtronic Bioporto, the Acute Kidney Injury Foundation, and Seastar; he receives grant funding for studies not related to
this project from Baxter, Nuwellis, Medtronic, and the National Institutes of Health; the other authors have indicated they have no potential conflicts of interest to disclose.

REFERENCES but chronically harmful programs. Evol 18. Thayyil S, Sheik S, Kempley ST, Sinha A.
1. Selewski DT, Charlton JR, Jetton JG, et Med Public Health. 2016; A gestation- and postnatal age-based
al. Neonatal acute kidney injury. Pediat- 2016(1):37–51 reference chart for assessing renal
rics. 2015;136(2). Available at: www. 10. Iacobelli S, Guignard JP. Maturation of function in extremely premature infants.
pediatrics.org/cgi/content/full/136/2/ glomerular filtration rate in neonates J Perinatol. 2008;28(3):226–229
e463 and infants: an overview. Pediatr Neph- 19. Askenazi DJ, Ambalavanan N, Goldstein
2. Askenazi DJ, Heagerty PJ, Schmicker RH, rol. 2021;36(6):1439–1446 SL. Acute kidney injury in critically ill
et al; PENUT Trial Consortium. Preva- 11. Hyink DP, Abrahamson DR. Origin of the newborns: what do we know? What do
lence of acute kidney injury (AKI) in glomerular vasculature in the develop- we need to learn? Pediatr Nephrol.
extremely low gestational age neonates ing kidney. Semin Nephrol. 1995;15(4): 2009;24(2):265–274
(ELGAN). Pediatr Nephrol. 2020;35(9): 300–314 20. Gupta C, Massaro AN, Ray PE. A new
1737–1748 12. Abitbol CL, Seeherunvong W, Galarza MG, approach to define acute kidney injury
3. Carmody JB, Swanson JR, Rhone ET, et al. Neonatal kidney size and function in term newborns with hypoxic ischemic
Charlton JR. Recognition and reporting in preterm infants: what is a true esti- encephalopathy. Pediatr Nephrol. 2016;
of AKI in very low birth weight infants. mate of glomerular filtration rate? 31(7):1167–1178
Clin J Am Soc Nephrol. 2014;9(12): J Pediatr. 2014;164(5):1026–1031.e2 21. Bezerra CT, Vaz Cunha LC, Liborio AB.
2036–2043 13. Su SW, Stonestreet BS. Core concepts: Defining reduced urine output in neona-
neonatal glomerular filtration rate. Neo- tal ICU: importance for mortality and
4. Jetton JG, Boohaker LJ, Sethi SK, et al;
reviews. 2010;11(12):e714–e721 acute kidney injury classification. Neph-
Neonatal Kidney Collaborative (NKC). Inci-
rol Dial Transplant. 2013;28(4):901–909
dence and outcomes of neonatal acute 14. Jetton JG, Guillet R, Askenazi DJ, et al;
kidney injury (AWAKEN): a multicentre, Neonatal Kidney Collaborative. Assess- 22. Bruel A, Roze JC, Flamant C, Simeoni U,
multinational, observational cohort ment of worldwide acute kidney injury Roussey-Kesler G, Allain-Launay E. Criti-
study. Lancet Child Adolesc Health. epidemiology in neonates: design of a cal serum creatinine values in very pre-
2017;1(3):184–194 retrospective cohort study. Front Pediatr. term newborns. PLoS One. 2013;8(12):
2016;4:68 e84892
5. Menon S, Broderick J, Munshi R, et al.
Kidney support in children using an 15. Kellum JA, Lameire N; KDIGO AKI Guide- 23. Askenazi D, Abitbol C, Boohaker L, et al;
ultrafiltration device: a multicenter, ret- line Work Group. Diagnosis, evaluation, Neonatal Kidney Collaborative. Optimiz-
rospective study. Clin J Am Soc Nephrol. and management of acute kidney injury: ing the AKI definition during first postna-
2019;14(10):1432–1440 a KDIGO summary (part 1). Crit Care. tal week using assessment of
2013;17(1):204 worldwide acute kidney injury epidemi-
6. Vidal E, Garzotto F, Parolin M, et al. Thera-
ology in neonates (AWAKEN) cohort.
peutic plasma exchange in neonates and 16. Zappitelli M, Ambalavanan N, Askenazi
Pediatr Res. 2019;85(3):329–338
infants: successful use of a miniaturized DJ, et al. Developing a neonatal acute
machine. Blood Purif. 2017;44(2):100–105 kidney injury research definition: a 24. Koralkar R, Ambalavanan N, Levitan EB,
report from the NIDDK neonatal AKI McGwin G, Goldstein S, Askenazi D. Acute
7. Chambers JM, Wingert RA. Advances in
workshop. Pediatr Res. 2017;82(4): kidney injury reduces survival in very
understanding vertebrate nephrogene- low birth weight infants. Pediatr Res.
569–573
sis. Tissue Barriers. 2020;8(4):1832844 2011;69(4):354–358
17. Stoops C, Stone S, Evans E, et al. Baby
8. Chevalier RL. Evolutionary nephrology. 25. Charlton JR, Boohaker L, Askenazi D, et
NINJA (Nephrotoxic Injury Negated by
Kidney Int Rep. 2017;2(3):302–317 Just-in-Time Action): reduction of neph- al; Neonatal Kidney Collaborative. Inci-
9. Straub RH, Schradin C. Chronic inflam- rotoxic medication-associated acute kid- dence and risk factors of early onset
matory systemic diseases: an evolution- ney injury in the neonatal intensive care neonatal AKI. Clin J Am Soc Nephrol.
ary trade-off between acutely beneficial unit. J Pediatr. 2019;215:223–228.e6 2019;14(2):184–195

10 STARR et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf
by Hospices Civils de Lyon user
26. Majed B, Bateman DA, Uy N, Lin F. Patent injury and the risk of chronic kidney dis- associated with higher morbidity in
ductus arteriosus is associated with ease. Kidney Int. 2017;92(3):751–756 pediatric cardiac surgery patients.
acute kidney injury in the preterm 36. Wong JH, Selewski DT, Yu S, et al. Severe Pediatr Crit Care Med. 2014;15(2):
infant. Pediatr Nephrol. 2019;34(6): acute kidney injury following stage 1 131–138
1129–1139 Norwood palliation: effect on outcomes 45. Li Y, Wang J, Bai Z, et al. Early fluid over-
27. Guillet R, Selewski DT, Griffin R, Rastogi and risk of severe acute kidney injury at load is associated with acute kidney
S, Askenazi DJ, D’Angio CT; Neonatal Kid- subsequent surgical stages. Pediatr Crit injury and PICU mortality in critically ill
ney Collaborative. Relationship of patent Care Med. 2016;17(7):615–623 children. Eur J Pediatr. 2016;175(1):
ductus arteriosus management with 37. Murphy HJ, Gien J, Sahay R, et al. Acute 39–48
neonatal AKI. J Perinatol. 2021;41(6): kidney injury, fluid overload, and renal 46. Mah KE, Hao S, Sutherland SM, et al. Fluid
1441–1447 replacement therapy differ by underly- overload independent of acute kidney
28. Charlton JR, Boohaker L, Askenazi D, et ing diagnosis in neonatal extracorporeal injury predicts poor outcomes in neo-
al; Neonatal Kidney Collaborative (NKC). support and impact mortality dispa- nates following congenital heart surgery.
Late onset neonatal acute kidney injury: rately [published online ahead of print Pediatr Nephrol. 2018;33(3):511–520
results from the AWAKEN Study. Pediatr January 18, 2021]. Blood Purif. doi: 47. Selewski DT, Cornell TT, Lombel RM, et
Res. 2019;85(3):339–348 10.1159/000512538 al. Weight-based determination of fluid
29. Polglase GR, Ong T, Hillman NH. Cardio- 38. Wu Y, Hua X, Yang G, Xiang B, Jiang X. overload status and mortality in pediat-
vascular alterations and multiorgan dys- Incidence, risk factors, and outcomes of ric intensive care unit patients requiring
function after birth asphyxia. Clin acute kidney injury in neonates after continuous renal replacement therapy.
Perinatol. 2016;43(3):469–483 surgical procedures. Pediatr Nephrol. Intensive Care Med. 2011;
2020;35(7):1341–1346 37(7):1166–1173
30. Bozkurt O, Yucesoy E. Acute kidney injury
in neonates with perinatal asphyxia 39. Garg PM, Britt AB, Ansari MAY, et al. 48. Sutherland SM, Zappitelli M, Alexander
receiving therapeutic hypothermia. Am J Severe acute kidney injury in neonates SR, et al. Fluid overload and mortality in
with necrotizing enterocolitis: risk fac- children receiving continuous renal
Perinatol. 2021;38(9):922–929
tors and outcomes [published online replacement therapy: the prospective
31. Kirkley MJ, Boohaker L, Griffin R, et al; ahead of print January 14, 2021]. pediatric continuous renal replacement
Neonatal Kidney Collaborative (NKC). Pediatr Res. doi: 10.1038/s41390-020- therapy registry. Am J Kidney Dis.
Acute kidney injury in neonatal encepha- 01320-6 2010;55(2):316–325
lopathy: an evaluation of the AWAKEN
40. Flori HR, Church G, Liu KD, Gildengorin 49. Valentine SL, Sapru A, Higgerson RA, et
database. Pediatr Nephrol. 2019;34(1):
G, Matthay MA. Positive fluid balance is al; Pediatric Acute Lung Injury and Sep-
169–176
associated with higher mortality and sis Investigator’s (PALISI) Network; Acute
32. Michniewicz B, Al Saad SR, Karbowski prolonged mechanical ventilation in Respiratory Distress Syndrome Clinical
LM, Gadzinowski J, Szymankiewicz M, pediatric patients with acute lung injury. Research Network (ARDSNet). Fluid bal-
Szpecht D. Organ complications of Crit Care Res Pract. 2011;2011:854142 ance in critically ill children with acute
infants with hypoxic ischemic encepha- 41. Gist KM, Selewski DT, Brinton J, Menon lung injury. Crit Care Med.
lopathy before therapeutic hypothermia. S, Goldstein SL, Basu RK. Assessment of 2012;40(10):2883–2889
Ther Hypothermia Temp Manag. the independent and synergistic effects 50. Hoste EA, Maitland K, Brudney CS, et al;
2021;11(1):58–63 of fluid overload and acute kidney injury ADQI XII Investigators Group. Four
33. Selewski DT, Jordan BK, Askenazi DJ, on outcomes of critically ill children. phases of intravenous fluid therapy: a
Dechert RE, Sarkar S. Acute kidney Pediatr Crit Care Med. 2020;21(2): conceptual model. Br J Anaesth.
injury in asphyxiated newborns treated 170–177 2014;113(5):740–747
with therapeutic hypothermia. J Pediatr. 42. Goldstein SL, Currier H, Graf JM, Cosio 51. Selewski DT, Akcan-Arikan A, Bonachea
2013;162(4):725–729.e1 CC, Brewer ED, Sachdeva R. Outcome in EM, et al; Neonatal Kidney Collaborative.
34. Fleming GM, Sahay R, Zappitelli M, et al. children receiving continuous venove- The impact of fluid balance on outcomes
The incidence of acute kidney injury and nous hemofiltration. Pediatrics. 2001; in critically ill near-term/term neonates:
its effect on neonatal and pediatric 107(6):1309–1312 a report from the AWAKEN study group.
extracorporeal membrane oxygenation 43. Gorga SM, Sahay RD, Askenazi DJ, et al. Pediatr Res. 2019;85(1):79–85
outcomes: a multicenter report from Fluid overload and fluid removal in pedi- 52. van Asperen Y, Brand PL, Bekhof J. Reli-
the kidney intervention during extracor- atric patients on extracorporeal mem- ability of the fluid balance in neonates.
poreal membrane oxygenation study brane oxygenation requiring continuous Acta Paediatr. 2012;101(5):479–483
group. Pediatr Crit Care Med. 2016; renal replacement therapy: a multicen- 53. Selewski DT, Gist KM, Nathan AT, et al;
17(12):1157–1169 ter retrospective cohort study. Pediatr Neonatal Kidney Collaborative. The
35. Madsen NL, Goldstein SL, Frøslev T, Nephrol. 2020;35(5):871–882 impact of fluid balance on outcomes in
Christiansen CF, Olsen M. Cardiac sur- 44. Hassinger AB, Wald EL, Goodman DM. premature neonates: a report from the
gery in patients with congenital heart Early postoperative fluid overload pre- AWAKEN study group. Pediatr Res.
disease is associated with acute kidney cedes acute kidney injury and is 2020;87(3):550–557

PEDIATRICS Volume 148, number 5, November 2021 11

Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf


by Hospices Civils de Lyon user
54. Bontant T, Matrot B, Abdoul H, et al. 65. Renganathan A, Warner BB, Tarr PI, Physiol Renal Physiol. 2019;317(4):
Assessing fluid balance in critically ill Dharnidharka VR. The progression of F865–F873
pediatric patients. Eur J Pediatr. 2015; serum cystatin C concentrations within 76. Baldelomar EJ, Charlton JR, Beeman SC,
174(1):133–137 the first month of life after preterm Bennett KM. Measuring rat kidney glo-
birth-a worldwide systematic review.
55. Paul IM, Schaefer EW, Miller JR, et al. merular number and size in vivo with
Pediatr Nephrol. 2021;36(7):1709–1718
Weight change nomograms for the first MRI. Am J Physiol Renal Physiol.
month after birth. Pediatrics. 2016; 66. Yang Y, Li SJ, Pan JJ, et al. Reference val- 2018;314(3):F399–F406
138(6):e20162625 ues for serum cystatin C in very low-
77. Rodrıguez MM, Gomez AH, Abitbol CL,
birthweight infants: from two centres of
56. Wadhawan R, Oh W, Perritt R, et al. Asso- Chandar JJ, Duara S, Zilleruelo GE. Histo-
China. J Paediatr Child Health. 2018;
ciation between early postnatal weight morphometric analysis of postnatal glo-
54(3):284–288
loss and death or BPD in small and merulogenesis in extremely preterm
appropriate for gestational age 67. Nakashima T, Inoue H, Fujiyoshi J, Matsu- infants. Pediatr Dev Pathol. 2004;7(1):
extremely low-birth-weight infants. moto N. Longitudinal analysis of serum 17–25
J Perinatol. 2007;27(6):359–364 cystatin C for estimating the glomerular
78. Charlton JR, Baldelomar EJ, deRonde KA,
filtration rate in preterm infants. Pediatr
57. Schmidt B, Roberts RS, Fanaroff A, et al; Nephrol. 2016;31(6):983–989 et al. Nephron loss detected by MRI
TIPP Investigators. Indomethacin prophy- following neonatal acute kidney injury
laxis, patent ductus arteriosus, and the 68. Treiber M, Pecovnik Balon B, Gorenjak in rabbits. Pediatr Res. 2020;87(7):
risk of bronchopulmonary dysplasia: fur- M. A new serum cystatin C formula for 1185–1192
ther analyses from the Trial of Indo- estimating glomerular filtration rate in
newborns. Pediatr Nephrol. 2015; 79. Harer MW, Chock VY. Renal tissue oxy-
methacin Prophylaxis in Preterms genation monitoring-an opportunity to
(TIPP). J Pediatr. 2006;148(6):730–734 30(8):1297–1305
improve kidney outcomes in the vulnera-
58. Oh W, Poindexter BB, Perritt R, et al; Neo- 69. Devarajan P. Neutrophil gelatinase-asso- ble neonatal population. Front Pediatr.
natal Research Network. Association ciated lipocalin: a promising biomarker 2020;8:241
for human acute kidney injury. Bio-
between fluid intake and weight loss 80. Hazle MA, Gajarski RJ, Aiyagari R, et al.
markers Med. 2010;4(2):265–280
during the first ten days of life and risk Urinary biomarkers and renal near-
of bronchopulmonary dysplasia in 70. Tanigasalam V, Bhat V, Adhisivam B, Srid-
infrared spectroscopy predict intensive
extremely low birth weight infants. har MG. Does therapeutic hypothermia
care unit outcomes after cardiac sur-
J Pediatr. 2005;147(6):786–790 reduce acute kidney injury among term
gery in infants younger than 6 months
neonates with perinatal asphyxia?–a
59. Askenazi DJ, Koralkar R, Hundley HE, of age. J Thorac Cardiovasc Surg.
randomized controlled trial. J Matern
Montesanti A, Patil N, Ambalavanan N. 2013;146(4):861–867.e1
Fetal Neonatal Med. 2016;29(15):
Fluid overload and mortality are associ- 81. Bonsante F, Ramful D, Binquet C, et al.
2545–2548
ated with acute kidney injury in sick Low renal oxygen saturation at near-
near-term/term neonate. Pediatr Neph- 71. Chen J, Sun Y, Wang S, et al. The effec-
infrared spectroscopy on the first day of
rol. 2013;28(4):661–666 tiveness of urinary TIMP-2 and IGFBP-7
in predicting acute kidney injury in criti- life is associated with developing acute
60. Selewski DT, Goldstein SL. The role of cally ill neonates. Pediatr Res. 2020; kidney injury in very preterm infants.
fluid overload in the prediction of out- 87(6):1052–1059 Neonatology. 2019;115(3):198–204
come in acute kidney injury. Pediatr 82. Dorum BA, Ozkan H, Cetinkaya M, Koksal
Nephrol. 2018;33(1):13–24 72. Fan W, Ankawi G, Zhang J, et al. Current
understanding and future directions in N. Regional oxygen saturation and acute
61. Askenazi DJ, Koralkar R, Patil N, Halloran the application of TIMP-2 and IGFBP7 in kidney injury in premature infants.
B, Ambalavanan N, Griffin R. Acute kid- AKI clinical practice. Clin Chem Lab Med. Pediatr Int. 2021;63(3):290–294
ney injury urine biomarkers in very low- 2019;57(5):567–576 83. Harer MW, Adegboro CO, Richard LJ,
birth-weight infants. Clin J Am Soc Neph-
73. Stojanovic VD, Barisic NA, Vuckovic NM, McAdams RM. Non-invasive continuous
rol. 2016;11(9):1527–1535
Doronjski AD, Peco Antic AE. Urinary kid- renal tissue oxygenation monitoring to
62. DeFreitas MJ, Seeherunvong W, Katsoufis ney injury molecule-1 rapid test predicts identify preterm neonates at risk for
CP, et al. Longitudinal patterns of urine bio- acute kidney injury in extremely low- acute kidney injury. Pediatr Nephrol.
markers in infants across gestational ages. birth-weight neonates. Pediatr Res. 2021;36(6):1617–1625
Pediatr Nephrol. 2016;31(7):1179–1188 2015;78(4):430–435 84. Chock VY, Frymoyer A, Yeh CG, Van
63. Wasung ME, Chawla LS, Madero M. Bio- 74. Charlton JR, Baldelomar EJ, Hyatt DM, Meurs KP. Renal saturation and acute
markers of renal function, which and Bennett KM. Nephron number and its kidney injury in neonates with hypoxic
when? Clin Chim Acta. 2015;438:350–357 determinants: a 2020 update. Pediatr ischemic encephalopathy undergoing
64. Chowdhary V, Vajpeyajula R, Jain M, et Nephrol. 2021;36(4):797–807 therapeutic hypothermia. J Pediatr.
al. Comparison of different definitions of 75. Baldelomar EJ, Charlton JR, deRonde KA, 2018;200:232–239.e1
acute kidney injury in extremely low Bennett KM. In vivo measurements of 85. Sanderson KR, Chang E, Bjornstad E, et
birth weight infants. Clin Exp Nephrol. kidney glomerular number and size in al. Albuminuria, hypertension, and
2018;22(1):117–125 healthy and Os/1 mice using MRI. Am J reduced kidney volumes in adolescents

12 STARR et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf
by Hospices Civils de Lyon user
born extremely premature. Front retrospective cohort of very low birth the very low birth weight infant. Neona-
Pediatr. 2020;8:230 weight neonates. J Pediatr. 2016;172: tology. 2016;110(4):307–312
86. Askenazi DJ, Heagerty PJ, Schmicker RH, 63–68.e1 106. Kraut EJ, Boohaker LJ, Askenazi DJ,
et al. The impact of erythropoietin on 95. Kaddourah A, Goldstein SL. Renal Fletcher J, Kent AL; Neonatal Kidney Col-
short- and long-term kidney-related out- replacement therapy in neonates. Clin laborative (NKC). Incidence of neonatal
comes in neonates of extremely low ges- Perinatol. 2014;41(3):517–527 hypertension from a large multicenter
tational age. Results of a multicenter, study [Assessment of Worldwide Acute
96. Harshman LA, Muff-Luett M, Neuberger
double-blind, placebo-controlled random- Kidney Injury Epidemiology in Neonates-
ML, et al. Peritoneal dialysis in an
ized clinical trial. J Pediatr. 2021; AWAKEN]. Pediatr Res. 2018;84(2):279–289
extremely low-birth-weight infant with
232:65–72.e7 acute kidney injury. Clin Kidney J. 107. Coca SG, Singanamala S, Parikh CR.
87. Pedersen KR, Ravn HB, Povlsen JV, 2014;7(6):582–585 Chronic kidney disease after acute kid-
Schmidt MR, Erlandsen EJ, Hjortdal VE. ney injury: a systematic review and
97. Munshi R, Lee-Son K, Hackbarth RM, et
Failure of remote ischemic precondition- meta-analysis. Kidney Int. 2012;81(5):
al. Clinical evaluation of the Prismaflex™
ing to reduce the risk of postoperative 442–448
HF 20 set and Prismaflex™ system 7.10
acute kidney injury in children undergo- 108. Greenberg JH, Coca S, Parikh CR. Long-
for acute continuous kidney replace-
ing operation for complex congenital term risk of chronic kidney disease and
ment therapy (CKRT) in children. Pediatr
heart disease: a randomized single-cen- mortality in children after acute kidney
Nephrol. 2020;35(12):2345–2352
ter study. J Thorac Cardiovasc Surg. injury: a systematic review. BMC Neph-
2012;143(3):576–583 98. Askenazi DJ. AWAKEN-ing a new frontier
rol. 2014;15:184
in neonatal nephrology. Front Pediatr.
88. Lomivorotov V, Kornilov I, Boboshko V, et 109. Chawla LS, Eggers PW, Star RA, Kimmel
2020;8:21
al. Effect of intraoperative dexametha- PL. Acute kidney injury and chronic kid-
sone on major complications and mor- 99. Garzotto F, Vidal E, Ricci Z, et al. Continu-
ney disease as interconnected syn-
tality among infants undergoing cardiac ous kidney replacement therapy in criti- dromes. N Engl J Med. 2014;371(1):
surgery: the DECISION randomized clini- cally ill neonates and infants: a 58–66
cal trial. JAMA. 2020;323(24):2485–2492 retrospective analysis of clinical results
with a dedicated device. Pediatr Neph- 110. Basile DP, Bonventre JV, Mehta R, et al;
89. Harer MW, Askenazi DJ, Boohaker LJ, et ADQI XIII Work Group. Progression after
rol. 2020;35(9):1699–1705
al; Neonatal Kidney Collaborative (NKC). AKI: understanding maladaptive repair
Association between early caffeine cit- 100. Faubel S, Shah PB. Immediate conse- processes to predict and identify thera-
rate administration and risk of acute quences of acute kidney injury: the peutic treatments. J Am Soc Nephrol.
kidney injury in preterm neonates: impact of traditional and nontraditional 2016;27(3):687–697
results from the AWAKEN study. JAMA complications on mortality in acute kid-
ney injury. Adv Chronic Kidney Dis. 111. Sutherland MR, Gubhaju L, Moore L, et
Pediatr. 2018;172(6):e180322
2016;23(3):179–185 al. Accelerated maturation and abnor-
90. Bellos I, Pandita A, Yachha M. Effective- mal morphology in the preterm neona-
ness of theophylline administration in 101. Basu RK, Wheeler DS. Kidney-lung cross- tal kidney. J Am Soc Nephrol.
neonates with perinatal asphyxia: a talk and acute kidney injury. Pediatr 2011;22(7):1365–1374
meta-analysis. J Matern Fetal Neonatal Nephrol. 2013;28(12):2239–2248
112. Abitbol CL, Bauer CR, Montane B,
Med. 2019;34(18):3080–3088 102. Grigoryev DN, Liu M, Hassoun HT, Chea- Chandar J, Duara S, Zilleruelo G. Long-
91. Mohamed TH, Klamer B, Mahan JD, dle C, Barnes KC, Rabb H. The local and term follow-up of extremely low birth
Spencer JD, Slaughter JL. Diuretic ther- systemic inflammatory transcriptome weight infants with neonatal renal fail-
apy and acute kidney injury in preterm after acute kidney injury. J Am Soc ure. Pediatr Nephrol. 2003;18(9):887–893
neonates and infants [published online Nephrol. 2008;19(3):547–558
113. Harer MW, Pope CF, Conaway MR,
ahead of print May 21, 2021]. Pediatr 103. Starr MC, Boohaker L, Eldredge LC, et Charlton JR. Follow-up of Acute Kidney
Nephrol. doi: 10.1007/s00467-021-05132-6 al; Neonatal Kidney Collaborative. Acute Injury in Neonates During Childhood
92. Kakajiwala A, Kim JY, Hughes JZ, et al. kidney injury and bronchopulmonary Years (FANCY): a prospective cohort
Lack of furosemide responsiveness pre- dysplasia in premature neonates born study. Pediatr Nephrol. 2017;32(6):
dicts acute kidney injury in infants after less than 32 weeks’ gestation. Am J 1067–1076
cardiac surgery. Ann Thorac Surg. Perinatol. 2020;37(3):341–348
114. Bruel A, Roze JC, Quere MP, et al. Renal
2017;104(4):1388–1394 104. Starr MC, Boohaker L, Eldredge LC, et outcome in children born preterm with
93. Chock VY, Cho SH, Frymoyer A. Aminoph- al; Neonatal Kidney Collaborative. Acute neonatal acute renal failure: IRENEO-a
ylline for renal protection in neonatal kidney injury is associated with poor prospective controlled study. Pediatr
hypoxic-ischemic encephalopathy in the lung outcomes in infants born $32 Nephrol. 2016;31(12):2365–2373
era of therapeutic hypothermia. Pediatr weeks of gestational age. Am J Perina- 115. Maqsood S, Fung N, Chowdhary V, Raina
Res. 2021;89(4):974–980 tol. 2020;37(2):231–240 R, Mhanna MJ. Outcome of extremely
94. Carmody JB, Harer MW, Denotti AR, 105. Stoops C, Sims B, Griffin R, Askenazi DJ. low birth weight infants with a history
Swanson JR, Charlton JR. Caffeine expo- Neonatal acute kidney injury and the of neonatal acute kidney injury. Pediatr
sure and risk of acute kidney injury in a risk of intraventricular hemorrhage in Nephrol. 2017;32(6):1035–1043

PEDIATRICS Volume 148, number 5, November 2021 13

Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf


by Hospices Civils de Lyon user
116. Askenazi DJ, Morgan C, Goldstein SL, et retrospective cohort study. J Perinatol. survey of neonatologists’ and neph-
al. Strategies to improve the understand- 2020;40(9):1301–1307 rologists’ perceptions and practice
ing of long-term renal consequences 118. Vincent K, Murphy HJ, Ross JR, management. Am J Perinatol.
after neonatal acute kidney injury. Pediatr Twombley KE. Acute kidney injury guide- 2018;35(1):1–9
Res. 2016;79(3):502–508 lines are associated with improved rec- 120. Nour I, Elmaghraby R, Shehata R, et al.
117. Starr MC, Kula A, Lieberman J, et al. The ognition and follow-up for neonatal Selective head cooling and acute
impact of increased awareness of acute patients. Adv Neonatal Care. 2020; kidney injury in neonates with
kidney injury in the neonatal intensive 20(4):269–275 hypoxic ischemic encephalopathy.
care unit on acute kidney injury inci- 119. Kent AL, Charlton JR, Guillet R, et al. J Neonatal Perinatal Med. 2020;13(1):
dence and reporting: results of a Neonatal acute kidney injury: a 21–30

14 STARR et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/148/5/e2021051220/1196738/peds_2021051220.pdf
by Hospices Civils de Lyon user

You might also like