You are on page 1of 6

Journal of Perinatology (2017) 00, 1–6

© 2017 Nature America, Inc., part of Springer Nature. All rights reserved 0743-8346/17
www.nature.com/jp

ORIGINAL ARTICLE
Erythropoietin monotherapy in perinatal asphyxia with
moderate to severe encephalopathy: a randomized
placebo-controlled trial
RR Malla1, R Asimi2, MA Teli1, F Shaheen3 and MA Bhat1

OBJECTIVE: Erythropoietin (EPO) is neuroprotective after asphyxia in animal studies. The efficacy and safety of EPO monotherapy in
term neonates with hypoxic ischemic encephalopathy (HIE) is uncertain.
STUDY DESIGN: Hundred term neonates with moderate or severe HIE were randomized by random permuted block algorithm to
receive either EPO 500 U kg− 1 per dose in 2 ml saline intravenously (50 neonates) on alternate days for a total of five doses with the
first dose given by 6 h of age (treatment group) or 2 ml of normal saline (50 neonates) similarly for a total of five doses (placebo
group) in a double-blind study. No hypothermia was given. The primary outcome was combined end point of death or moderate or
severe disability at mean age of 19 months (s.d., 0.61).
RESULTS: Death or moderate or severe disability occurred in 40% of neonates in the treatment group vs 70% in the placebo group
(risk ratio, 0.57; 95% confidence interval (CI) 0.38 to 0.85; P = 0.003). Death occurred in 16% of patients in both the groups (risk ratio,
1.0; 95% CI 0.33 to 2.9; P = 0.61). The risk of cerebral palsy was lower among survivors in the treatment group (risk ratio, 0.52; 95% CI
0.25 to 1.03; P = 0.04) and lesser number of babies were on anticonvulsants at assessment (risk ratio, 0.47; 95% CI 0.20 to 1.01;
P = 0.03). Neonatal brain magnetic resonance imaging showed more abnormalities in the placebo group (relative risk, 0.66; 95% CI
0.42 to 1.03; P = 0.04)). Improvement in other neurological outcomes was not significant.
CONCLUSION: EPO monotherapy reduces the risk of death or disability in term neonates with moderate or severe encephalopathy.
Journal of Perinatology advance online publication, 9 March 2017; doi:10.1038/jp.2017.17

INTRODUCTION encouraging.17,18 These studies suggest that EPO may be used


Hypoxic ischemic encephalopathy (HIE) is associated with signifi- with hypothermia for neuroprotection or may be used as an
cant morbidity and mortality during neonatal period and neurode- acceptable alternative in resource poor settings where standard
velopmental disability in the long term. Patients with moderate HIE cooling machines are not available. As 23% of neonatal deaths
have a 10% risk of death and 30% risk of disability among survivors. occur as a consequence or related to HIE and most of these occur
Among patients with severe HIE, 60% patients die and almost all in regions where cooling is not available, early treatment with EPO
survivors develop disability.1–3 Therapeutic hypothermia when may provide a much needed approach to improve outcomes.
initiated within first 6 h of birth has been shown to improve the Although most studies of additional neuroprotective therapies for
immediate as well as long-term outcome in HIE.4–7 However clinical HIE focus on testing an adjuvant therapy to be given in
trials suggest that around 24 to 38% of infants who receive conjunction with hypothermia, this study is asking whether a
different therapy altogether is effective, that can be used in low-
hypothermia die and another 35 to 41% of patients develop
resource settings where hypothermia is unavailable or not
developmental disability at 18 to 22 months of age.4,8 Also
available for all babies.
hypothermia is most beneficial to babies with moderate encepha-
lopathy and meta-analysis show minor benefit to babies with
severe HIE.6 So there is need for other modalities that could also MATERIALS AND METHODS
have neuroprotective as well as neurorestorative properties. This was a prospective longitudinal study conducted in the neonatal
Recombinant human erythropoietin (EPO) has been extensively intensive care unit of the Sheri Kashmir Institute of Medical Sciences
studied as a neuroprotective agent in animal models. EPO and (SKIMS), Srinagar, Kashmir, which is a tertiary-care hospital in northern
erythropoietin receptor are expressed by various cells in the brain India from December 2012 to November 2015. The infants enrolled in the
that include neuronal progenitor cells, subsets of mature neurons, study were born in the obstetric department of SKIMS, Jhelum Valley
oligodendrocytes, astrocytes and microglia.9–14 Hypoxic ischemia Medical College and Lalded hospital. The total number of babies born in
the obstetric unit of SKIMS and Jhelum Valley Medical College is 10 000 per
of the brain leads to increased expression of EPO and
year, whereas at Lalded hospital it is 22 000 per year. All infants were
erythropoietin receptor in neurons mediated by hypoxia inducible enrolled after parental consent (written and verbal format) and the study
factor, which is an endogenous neuroprotective mechanism.15,16 was approved by the Institutional Ethics Committee (IEC).
Few studies have evaluated the neuroprotective role of EPO in All neonates who were ⩾ 37 weeks of gestation, o6 h of age and had
term neonates with perinatal asphyxia and the results were evidence of moderate or severe HIE were eligible for inclusion in the study.

1
Department of Paediatrics, Sheri Kashmir Institute of Medical Sciences, Srinagar, India; 2Department of Neurology, Sheri Kashmir Institute of Medical Sciences, Srinagar, India and
3
Department of Radiodiagnosis and Imaging, Sheri Kashmir Institute of Medical Sciences, Srinagar, India. Correspondence: Professor MA Bhat, Department of Paediatrics, Sheri
Kashmir Institute of Medical Sciences, Srinagar 190011, India.
Email: mbhat47@rediffmail.com
Received 26 July 2016; revised 7 January 2017; accepted 17 January 2017
Erythropoietin neuroprotection in perinatal asphyxia
RR Malla et al
2
Perinatal asphyxia was defined as 10 min Apgar score o 5 together with to move.21 Developmental examination was done by Bayley Scale of Infant
evidence of two of the following three criteria and evidence of moderate Development II on which the standardization mean (± s.d.) is 100 ±15.22
or severe neonatal encephalopathy. Severe disability was defined as GMFCS grade of level 3 to 5, hearing
impairment requiring hearing aids, bilateral cortical visual impairment with
1. History of fetal distress (late deceleration or loss of beat-to-beat no useful vision or Bayley Mental Development Index Score o70.
variability or fetal bradycardia or meconium-stained amniotic fluid). Moderate disability was defined as Bayley Mental Developmental Index
2. Need for immediate neonatal ventilation with a bag and mask or Score between 70 and 84 and any one of the following criteria: GMFCS
through endotracheal intubation for ⩾ 10 min after delivery. grade of level 2, hearing impairment with no amplification or persistent
3. Base deficit of ⩾ 16 mEq l − 1 and/or pH 7.0 or less in cord blood or seizure disorder.
admission arterial blood samples taken within the first hour after birth. Adverse outcomes included intracranial hemorrhage, pulmonary
hemorrhage, pulmonary hypertension, persistent hypotension, prolonged
Moderate or severe neonatal encephalopathy in these neonates was coagulation time, culture-proven sepsis, necrotizing enterocolitis, throm-
diagnosed by certified examiners when one sign was present in 43 of the bocytopenia, major venous thrombosis, renal failure needing dialysis,
pulmonary air leaks, duration of hospitalization.
following six categories:19
Secondary outcomes at 19 months included disability, Bayley Mental
and Psychomotor Developmental Index (mean score 100 ± 15), cerebral
(1) Level of consciousness: lethargy (moderate HIE) or stupor or coma palsy, cortical visual impairment, hearing loss, seizures needing
(severe HIE). anticonvulsants.
(2) Spontaneous activity: decreased (moderate HIE) or absent (severe HIE).
(3) Tone: hypotonia (moderate HIE) or flaccidity (severe HIE).
(4) Posture: distal flexion (moderate HIE) or decerebrate state (severe HIE). Statistical analysis
(5) Primitive reflexes: suck weak (moderate HIE) or absent (severe HIE) or Sample size of 50 infants in each group was based on a two-tailed type 1
moro reflex incomplete (moderate HIE) or absent (severe HIE). error rate of 0.05%, a statistical power of 80 and 10% loss to the follow-up
(6) Autonomic nervous system: pupils constricted (moderate HIE) or and an incidence of death or disability in the control group of 70% and a
deviated, dilated or non-reactive to light (severe HIE), bradycardia reduction by 50% in the intervention group. The data were analyzed by
(moderate HIE) or variable heart rate (severe HIE), or periodic breathing Fisher’s exact tests for the categorical variables and with t-tests for the
(moderate HIE) or apnea (severe HIE). continuous variables. The data for the primary and secondary outcomes
were analyzed by the Mantel–Haenszel test.
On duty neonatologists (six senior residents with post graduation in
pediatrics ) acted as certified examiners and were certified when there was
concordance in three of the examinations with that of the lead instructors RESULTS
regarding the stage of HIE. From December 2012 to November 2015, 293 neonates with
Babies with congenital anomalies, chromosomal abnormalities, con- diagnosis of HIE were screened. Out of these, 163 did not meet the
genital infections, severe intrauterine growth retardation (o2 s.d. below
inclusion criteria (no moderate or severe encephalopathy) and 20
the mean) and inborn errors of metabolism were excluded from our study.
The patients were randomized into treatment and placebo group, and patients met criteria for exclusion. Of the remaining 110 patients,
randomization was done by random permuted block algorithm with block 10 patients were excluded because parents refused to participate
size of two and four. Treatment group received recombinant human EPO (Box1). Remaining 100 patients (50 in the treatment group and 50
within 6 h after birth at a dose of 500 U kg − 1 intravenously on alternate in the placebo group) were enrolled into the study. Baseline
days for a total of five doses. It was diluted in 2 ml of normal saline and characteristics of the infants were broadly similar between the two
administered intravenously over a period of 1 min. Placebo group received groups (Table 1).
2 ml of normal saline on the same schedule. The investigators, caregivers
and families were blinded to the assignment of patients (double blind).
All the infants were nursed on servo-controlled open care beds, with Adverse outcomes
skin temperature maintained at 36.5°C. No hypothermia was given. The incidence of adverse outcomes was similar between the two
Electroencephalography monitoring was done at admission and subse- groups (Table 2). Hypotension, hepatic dysfunction, prolonged
quently at 10 to 14 days of life depending upon the condition of the baby. coagulation and necrotizing enterocolitis were similar between
All electroencephalography's were reported by neurologists blinded to the the two groups. Although red blood cell indices (hemoglobin, red
assignment of patients. blood cell count and reticulocyte count) were elevated in the
During the initial 72 h of life, heart rate, respiratory rate, blood pressure
and oxygen saturation was monitored continuously. HIE scoring was done
treatment group on day 10 of life, these normalized by 1 month of
at admission and every 12 h during the initial 7 days and subsequently on age and no complications were seen. There were no patients with
day 10 and day 14 according to the Thompson’s HIE score.20 Mean HIE hypertension, polycythemia or thrombosis secondary to EPO.
score for each baby was calculated daily by taking an average of all scores
every 24 h. Arterial blood gases, electrolyte measurements, hemogram, Primary outcome at 19 months
renal function tests and liver function tests were performed daily for the
first 5 days or earlier depending upon the condition of the baby.
In the treatment group, 8 patients died (6 in the neonatal period
Hemogram was repeated on day 10, 1 and 3 months of life. All neonates and 2 during follow-up) and 12 patients survived with severe or
were subjected to magnetic resonance imaging (MRI) between tenth and moderate disability, whereas in the placebo group 8 patients died
fourteenth day of life or as permitted by the condition of the baby. All (6 in the neonatal period and 2 during follow-up) and 27 patients
MRI's were reported by neuroradiologist blinded to the assignment of survived with severe or moderate disability (Table 3; relative risk,
patients. 0.57; 95% confidence interval (CI), 0.38 to 0.85; number needed to
treat 4).
Outcome
The primary outcome was death or moderate or severe disability at mean Secondary outcome at 19 months
age of 19 months (s.d., 0.61). Data regarding growth, brainstem auditory- Outcome was significantly better in the treatment group in many
evoked response, vision were obtained. Neuromotor disability was based of the secondary outcome parameters (Table 3).There was less risk
on the presence of cerebral palsy and functional disability on the basis of
of death or disability among patients with moderate encephalo-
Gross Motor Function Classification (GMFCS) system where level 1 includes
children who walk independently with some gait abnormalities; level 2 pathy in the treatment group (relative risk, 0.33; 95% CI 0.12 to
includes those who are unable to walk but who can sit, pull to standing 0.79; P = 0.004). Survival without neurological abnormality was
and cruise; level 3 includes those who are unable to walk or crawl, use significantly increased in the treatment group (71%) than in the
hands for sitting support; level 4 includes those for whom support is placebo group (30%) (relative risk, 0.65; 95% CI 0.45 to 0.94;
needed for sitting; and level 5 includes those who require adult assistance P = 0.016). Patients in the treatment group had lower risk of

Journal of Perinatology (2017), 1 – 6 © 2017 Nature America, Inc., part of Springer Nature.
Erythropoietin neuroprotection in perinatal asphyxia
RR Malla et al
3
cerebral palsy (relative risk, 0.52; 95% CI 0.25 to 1.03; P = 0.04) than ratio, 0.47; 95% CI 0.20 to 1.01; P = 0.03). There was no effect on
patients in the placebo group. Also lesser number of babies were rest of the parameters between the two groups.
on anticonvulsants at assessment in the treatment group (risk On subgroup analysis more patients with severe encephalo-
pathy (34/50, 68%) died or had disability than those with
moderate encephalopathy (21/50, 42%) (relative risk, 0.55; 95%
CI 0.33 to 0.90; P = 0.009)
Table 1. Baseline characteristics of the infants

Variable Treatment Placebo group P-value Neonatal outcomes


group During neonatal period, 12% patients died in each group.
Abnormal electroencephalography at discharge was seen in 33%
(n = 50) (n = 50) ( o0.05 sig.) of patients in the treatment group vs 66% of patients in the
placebo group (relative risk, 0.81; 95% CI 0.51 to 0.32; P = 0.001).
Males 28 (56%) 31 (62%) 0.46 Abnormalities on MRI were seen more in the placebo group
Birth weight (grams) 2902 ± 444 3136 ± 660 0.9
(Table 2; relative risk, 0.66; 95% CI 0.42 to 1.03; P = 0.04). The
(mean ± s.d.)
Gestational age (weeks) 39.4 ± 1.23 39.7 ± 0.78 0.30 abnormalities included injury to the subcortical areas (posterior
Mode of delivery limb of internal capsule, thalamus, basal ganglia), cortical injuries
Vaginal 12 (24%) 16 (32%) 0.36 and cerebellar injuries.
Emergency cesarean 38 (76%) 34 (68%)

Meconium-stained 32 (64%) 22 (54.2%) 0.44 DISCUSSION


liquor Our study shows that treatment with EPO caused a significant
Apgar score 4.9 ± 1.3 4.7 ± 0.8 0.43 reduction in the risk of primary outcome, the combined rates of
(mean ± s.d.) (10 min)
Resuscitation required at 46 (92%) 45 (90%) 0.9 death or moderate or severe encephalopathy.
10 min The immediate neuroprotective effects of EPO include
Blood gases (cord blood or within first hour after birth) decreased nitric oxide production, activation of antioxidant
pH (mean ± s.d.) 7.00 ± 0.13 7.0 ± 0.11 0.26 enzymes, reduction of glutamate toxicity, inhibition of lipid
BE (mean ± s.d.) 18.9 ± 4.3 17.7 ± 4.0 0.21 peroxidation and reduction of inflammation, whereas the long-
term neuroprotective effects include generation of neuronal anti-
Mean HIE score (± s.d.) 12.5 ±2.9 12.5 ± 3.5 0.14 apoptotic mechanisms, stimulation of angiogenesis and modula-
Age at first dose 1.12 (1.5– 1.5 (2–3.5) 0.5
(interquartile range, 2.62)
tion of neurogenesis.23
25th–75th percentile) Under normal circumstances only 1 to 2% of circulating EPO
Mean arterial blood 45.3 ± 5.0 45.5 ± 4.6 0.861 crosses blood–brain barrier mainly by passive diffusion.24,25
pressure at admission However in the setting of hypoxic ischemia, there is an increased
(mean ± s.d.) permeability of the blood–brain barrier and doses of 300 to 500
Stage of HIE U kg − 1 are associated with a significant rise in cerebrospinal fluid
Moderate 24 (48%) 26 (52%) 0.65 EPO levels.26,27 Zhu et al.17 in their study of 83 infants (30 received
Severe 26 (52%) 24 (48%) 0.65
EPO at a dosage of 500 IU kg −1 per day and the rest at a dosage of
Abbreviations: BE, Base excess; HIE, hypoxic ischemic encephalopathy. 300 IU kg − 1 per day) reported EPO to be safe in all the treated
infants. High doses of EPO given subcutaneously in neonatal rat

Table 2. Status during the hospital stay and at discharge

Variable Treatment group Placebo group Relative risk P-value

N = 50(%) N = 50(%) (95% CI)

Oliguria 21 (42%) 30 (60%) 0.70 (0.45–1.07) 0.07


Renal failure 20 (40%) 28 (56%) 0.71 (0.45–1.12) 0.17
Hypotension requiring vasopressors 30 (60%) 33 (66%) 0.90 (0.65–1.26) 0.10
Hepatic dysfunction 18 (36%) 21 (42%) 0.85 (0.49–1.4) 0.54
Necrotizing enterocolitis 1 (2%) 4 (8%) 0.25 (0.01–2.27) 0.16
Disseminated intravascular coagulation 15 (30%) 18 (36%) 0.83 (0.44–1.54) 0.78
Ventilated 18 (36%) 17 (34%) 1.05 (0.58–1.91) 0.83
Death (neonatal period) 6 (12%) 6 (12%) 1.0 (0.33–2.9) 1.0
Day of initiation of oral feeds (mean ± s.d.) 6.0 ± 4.3 9.7 ± 5.1 0.006
Full oral feeds (mean ± s.d. days) 7.4 ± 4.8 11.3 ± 5.3 0.008
Mean duration of hospital stay (mean ± s.d.) 9.7 ± 6.9 13.5 ± 8.1 0.04
Hemoglobin on day 10 (± s.d.) 17.4 ±1.9 15.7 ± 2.2 0.004
Red blood cell count on day 10 (± s.d.) 5.2 ±0.45 5.2 ± 0.45 0.014
Reticulocyte count on day 10 (± s.d.) 4.1 ±0.5 3.2 ± 0.5 0.001
MRI abnormalities 20 (40%) 30 (60%) 0.66 (0.42–1.03) 0.04
Status at discharge
Gavage feeding 5 (10%) 6 (12%) 0.83 (0.23–2.9) 0.75
Abnormal EEG 17 (34%) 33 (66%) 0.81 (0.51–0.32) 0.001
Seizures requiring treatment 13 (26%) 28 (57%) 0.46 (0.25–0.80) 0.002
Abbreviations: CI, confidence interval; EEG, electroencephalography; MRI, magnetic resonance imaging.

© 2017 Nature America, Inc., part of Springer Nature. Journal of Perinatology (2017), 1 – 6
Erythropoietin neuroprotection in perinatal asphyxia
RR Malla et al
4
Table 3. Outcome at 19 months of age

Variable Treatment group Placebo group Relative risk P-value (95% CI)

Primary outcome
Death or moderate or severe disability 20/50 (40%) 35/50 (70%) 0.57 (0.38–0.85) 0.003

Secondary outcome
Death 8/50 (16%) 8/50 (16%) 1 (0.33–2.9) 0.61
Death or disability
Among patients with moderate encephalopathy 5/24 (21%) 16/26 (61%) 0.33 (0.12–0.79) 0.004
Among patients with severe encephalopathy 15/26 (57%) 19/24 (79%) 0.72 (0.51–1.3) 0.10
Neurodevelopmental abnormalities 12/42 (29%) 27/42 (64%) 0.44 (0.25–0.76) 0.001
Survival without neurological abnormalities 30/42 (71%) 15/42 (35%) 0.65 (0.45–0.94) 0.01
Cerebral palsy 10/42 (23%) 19/42 (45%) 0.52 (0.25–1.03) 0.04
Hearing impairment 2/42 (4.7%) 2/42 (4.7%) 1 (0.33–2.9) 1.0
Seizures requiring anticonvulsants at the time of assessment 8/42 (19%) 17/42 (43%) 0.47 (0.20–1.01) 0.03

Bayley Mental Development Index Score


o70 8/40 (20%) 15/40 (37%) 0.53 (0.22–1.18) 0.08
70–84 5/40 (12%) 6/40 (15%) 0.83 (0.23–2.90) 0.74
485 27/40 (67%) 19/40 (47%) 0.62 (0.34–1.10) 0.07

Bayley Psychomotor Development Index Score


o70 9/40 (23%) 15/40 (37%) 0.60 (0.26–1.28) 0.14
70–84 4/40 (10%) 5/40 (13%) 0.80 (0.18–3.2) 0.72
485 27/40 (67%) 20/40 (50%) 0.65 (0.35–1.17) 0.11
Abbreviation: CI, confidence interval. For Bayley scale of infant development testing, data for two patients in both the groups were not available.

models (2500 and 5000 units) have been found to be safe and years.33 A study of 45 term infants comparing single-dose EPO
improved the development of hypoxia-exposed newborn rats and alone on day 0 with 72 h therapeutic hypothermia alone for
prevented learning impairment and dopamine neuron loss in treatment of NE found superior protection in the hypothermia
these rats.27 Wu et al.28 have reported EPO dose of 1000 U kg − 1 to group.34 In a recent study of high-dose EPO and hypothermia for
produce plasma levels in neonates that are neuroprotective in HIE, Wu et al.35 demonstrated reduced severity of brain injury on
animals. However we used 500U kg − 1 of EPO keeping in view the neonatal MRI and improved short-term motor outcomes in those
safety profile seen with studies by Zhu et al. and the side effects patients who received EPO as an adjunctive therapy to
seen in adults with high doses of EPO. hypothermia. Neonatal MRI in our study showed more normal
There was reduced risk of death or disability in the treatment brain MRI in the treatment group than in the placebo group
group than in the placebo group in our study (relative risk, 0.57; consistent with above results.
95% CI 0.38 to 0.85; P = 0.003). Zhu et al.17 in their study also We could not demonstrate a beneficial effect of EPO in patients
reported reduced risk of death or disability in the EPO group than with severe encephalopathy as death or disability was more in
in the control group (relative risk 0.62; 95% CI 0.41 to 0.94; these neonates (relative risk, 0.55; 95% CI 0.33 to 0.90; P = 0.009).
P = 0.017). Death or disability rate in the placebo group in our This could be because of the severity of the initial insult or
study was 70% vs 43.8% in the study by Zhu et al. This was because of inadequate neuroprotection with our dose of EPO as
because of lesser number of patients with severe HIE in the study Wu et al.28 have demonstrated EPO dose of 1000 U kg − 1 to
by Zhu et al. than in our study (26% vs 50%). However the death produce plasma levels in neonates that are neuroprotective in
or disability rate of 70% in the placebo group in our study is animals.
similar to the death or disability rate of 66.6% reported by the Expect for mild increase in hemoglobin and mean red blood cell
other studies.4,8 volume (which normalized in the neonatal period), no side effects
There was an improvement in survival without neurological of EPO were documented in our patients. Studies of neuropro-
abnormality (relative risk 0.65; 95% CI 0.45 to 0.94; P = 0.01) in the tective role of EPO in term and preterm neonates performed to
treatment group in our study. Also the risk of developing cerebral study appropriate dosing, effectiveness and safety have also not
palsy was less in the treatment group (relative risk, 0.52; 95% CI seen any side effects like polycythemia, thrombosis and hyperten-
0.25 to 1.03; P = 0.04). Various animal studies have explored the sion as are seen in adults28,36
mechanism through which EPO exerts its neuroprotective effect. The strength of our study is that this is the first study where a
In their study of neonatal rats with focal cerebral ischemia, Chang consistent dose of 500 U kg − 1 of EPO was given to the neonates
et al.29 noted markedly preserved hemispheric volume in the EPO- in the treatment group within 6 h of life. Also MRI and
treated group. During the acute post-injury period, EPO had anti electroencephalography were done in all the patients in the
inflammatory and anti-apoptotic roles, and during recovery it has neonatal period. Follow-up was good and no patient was lost
neurogenic and vasculogenic effects.30,31 In human adults with during the follow-up. However there are many limitations. We
stroke, EPO was shown to reduce infarction size slightly and to could study only 100 patients over a period of 2 years being a
improve clinical outcomes slightly.32 single-center study. Better sample size as used in various
EPO has also shown promising results when combined with hypothermia trials would have been more beneficial. Also lower
hypothermia for HIE. Follow-up of 22 infants enrolled in a phase I dose of EPO could be one of the reasons for not demonstrating
clinical trial of EPO-augmented hypothermia (no comparison beneficial effect in neonates with severe encephalopathy. As only
group) for treatment of neonatal encephalopathy found no deaths severe disabilities will be picked up by 19 months of age and
and only one infant with moderate–severe disability at age 2 minor impairments might go unrecognized, evaluation at 6 to 7

Journal of Perinatology (2017), 1 – 6 © 2017 Nature America, Inc., part of Springer Nature.
Erythropoietin neuroprotection in perinatal asphyxia
RR Malla et al
5
2 Robertson CMT, Finer NN, Grace MGA. School performance of survivors of neo-
Box 1 Method of enrollment of study patients
natal encephalopathy associated with birth asphyxia at term. J Pediatr 1989; 114:
753–760.
Assessed for eligibility 3 Busto R, Dietrich WD, Globus MYT, Valdes I, Scheinberg P, Ginsberg MD. Small
N = 293 differences in intraischemic brain temperature critically determine the extent of
Not meeting inclusion criteria (n = 163) ischemic neuronal injury. J Cereb Blood Flow Metab 1987; 7: 729–738.
(no moderate or severe encephalopathy) 4 Shankaran S, Laptook AR, Ehrenkranz RA, Tyson JE, McDonald SA, Donovan EF
Exclusion criteria (n = 20) et al. Whole-body hypothermia for neonates with hypoxic-ischemic encephalo-
pathy. N Engl J Med 2005; 353: 1574–1584.
Refused to participate
5 Jacobs SE, Morley CJ, Inder TE, Stewart MJ, Smith KR, McNamara PJ. Whole body
(n = 10) hypothermia for term and near term newborns with hypoxic-ischemic encepha-
Randomized lopathy: a randomized controlled trial. Arch Pediatr Adolesc Med 2011; 165:
(n = 100) 692–700.
Allocated to intervention: Allocated to intervention 6 Edwards AD, Brocklehurst P, Gunn AJ, Halliday H, Juszczak E, Levene M et al.
(Erythropoietin): (Placebo) Neurological outcomes at 18 months of age after moderate hypothermia for
(n = 50): (n = 50) perinatal hypoxic ischaemic encephalopathy: synthesis and meta-analysis of
Received allocated intervention: Received allocated intervention trial data. BMJ2010; 340: c363.
(n = 50): (n = 50) 7 Pappas A, Shankaran S, McDonald SA, Vohr BR, Hintz SR, Ehrenkranz RA et al.
Lost to follow-up: Lost to follow-up Cognitive outcome after neonatal encephalopathy. Pediatrics 2015; 135: e624–e638.
8 Azzopardi DV, Strohm B, Edwards AD, Dyet L, Halliday HL, Juszczak E et al.
(n = 0): (n = 0)
Moderate hypothermia to treat perinatal asphyxial encephalopathy. N Engl J Med
Final analysis: Final analysis 2009; 361: 1349–1358.
(n = 50): (n = 50) 9 Wu YW, Gonzalez FF. Erythropoietin: a novel therapy for hypoxic-ischaemic
encephalopathy? Dev Med Child Neurol 2015; 573: 34–39.
10 Wang L, Zhang Z, Wang Y, Zhang R, Chopp M. Treatment of stroke with
erythropoietin enhances neurogenesis and angiogenesis and improves neurolo-
years of age is needed to assess for improvement in other gical function in rats. Stroke 2004; 35: 1732–1737.
parameters. 11 Bernaudin M, Marti HH, Roussel S, Divoux D, Nouvelot A, MacKenzie ET et al. A
In conclusion, our study confirms that treatment with EPO potential role for erythropoietin in focal permanent cerebral ischemia in mice.
during the first 6 h of life in neonates with moderate to severe J Cereb Blood Flow Metab 1999; 19: 643–651.
encephalopathy improves the combined outcome of death or 12 Mu D, Chang YS, Vexler ZS, Ferriero DM. Hypoxia-inducible factor 1 alpha and
moderate or severe disability at 19 months of age. In settings erythropoietin upregulation with deferoxamine salvage after neonatal stroke. Exp
where hypothermia is unavailable or not available for all babies, Neurol 2005; 195: 407–415.
13 Wallach I, Zhang J, Hartmann A, van Landeghem FK, Ivanova A, Klar M et al.
EPO may be a much needed neuroprotective therapy as it has a
Erythropoietin-receptor gene regulation in neuronal cells. Pediatr Res 2009; 65:
low number needed to treat and is relatively easy to administer. At 619–624.
centers where hypothermia is available, it is used as an add on 14 Sugawa M, Sakurai Y, Ishikawa-Ieda Y, Suzuki H, Asou H. Effects of erythropoietin
therapy due to its neuroprotective properties. However more on glial cell development; oligodendrocyte maturation and astrocyte prolifera-
multicentric trials are needed to corroborate the results. tion. Neurosci Res 2002; 44: 391–403.
15 Nagai A, Nakagawa E, Choi HB, Hatori K, Kobayashi S, Kim SU. Erythropoietin
and erythropoietin receptors in human CNS neurons, astrocytes, microglia,
and oligodendrocytes grown in culture. J Neuropathol Exp Neurol 2001; 60:
What is known about topic
386–392.
● The efficacy and safety of erythropoietin in term neonates with
16 Bernaudin M, Nedelec AS, Divoux D, MacKenzie ET, Petit E, Schumann-Bard P.
perinatal asphyxia is uncertain.
Normobaric hypoxia induces tolerance to focal permanent cerebral ischemia in
association with an increased expression of hypoxia-inducible factor-1 and its
What this study adds
● Erythropoietin reduces the risk of death or disability in term
target genes, erythropoietin and VEGF, in the adult mouse brain. J Cereb Blood
Flow Metab 2002; 22: 393–403.
neonates with perinatal asphyxia with moderate to severe disability.
17 Zhu C, Kang W, Xu F, Cheng X, Zhang Z, Jia L et al. Erythropoietin improved
neurologic outcomes in newborns with hypoxic- ischemic encephalopathy.
Pediatrics 2009; 124: e218–e226.
18 Elmahdy H, El-Mashad AR, El-Bahrawy H, El-Gohary T, El-Barbary A, Aly H. Human
CONFLICT OF INTEREST recombinant erythropoietin in asphyxia neonatorum: pilot trial. Pediatrics 2010;
The authors declare no conflict of interest. 125: e1135–e1142.
19 Shalak LF, Laptook AR, Velaphi SC, Perlman JM. Amplitude-integrated
electroencephalography coupled with an early neurologic examination enhan-
ACKNOWLEDGEMENTS ces prediction of term infants at risk for persistent encephalopathy. Pediatrics
We, Dr Rahid and Dr Mushtaq, had full access to all of the data in the study and take 2003; 111(2): 351–357.
responsibility for the integrity of the data and the accuracy of the data analysis. 20 Thompson CM, Puterman AS, Linley LL, Hann FM, van der Elst CW, Molteno CD
et al. The value of a scoring system for hypoxic ischaemic encephalopathy in
predicting neurodevelopmental outcome. Acta Paediatr 1997; 86(7): 757–761.
AUTHOR CONTRIBUTIONS 21 Palisano RJ, Hanna SE, Rosenbaum PL, Russell DJ, Walter SD, Wood EP et al.
Dr Mushtaq conceptualized and designed the study, drafted the initial Validation of a model of gross motor function for children with cerebral palsy.
manuscript and approved the final manuscript as submitted. Drs Rahid, Rouf Phys Ther 2000; 80: 974–985.
and Manzoor carried out the initial analyses, reviewed and revised the 22 Bayley N. Bayley Scales of Infant Development–II. Psychological Corporation: San
Antonio, Texas, 1993.
manuscript, and approved the final manuscript as submitted. Dr Feroz
23 Kumral A, Tüzün F, Oner MG, Genç S, Duman N, Ozkan H. Erythropoietin in
coordinated and supervised data collection, critically reviewed the manuscript
neonatal brain protection: the past, the present and the future. Brain Dev 2011;
and approved the final manuscript as submitted. 33: 632–643.
24 Brines ML, Ghezzi P, Keenan S, Agnello D, de Lanerolle NC, Cerami C et al.
Erythropoietin crosses the blood–brain barrier to protect against experimental
REFERENCES brain injury. Proc Natl Acad Sci USA 2000; 97: 10526–10531.
1 Shankaran S, Woldt E, Koepke T, Bedard MP, Nandayal R. Acute neonatal 25 Juul SE, McPherson RJ, Farrell FX, Jolliffe L, Ness DJ, Gleason CA. Erythropoietin
morbidity and long-term central nervous system sequelae of perinatal asphyxia in concentrations in cerebrospinal fluid of nonhuman primates and fetal sheep
term infants. Early Hum Dev 1991; 25: 135–148. following high-dose recombinant erythropoietin. Biol Neonate 2004; 85: 138–144.

© 2017 Nature America, Inc., part of Springer Nature. Journal of Perinatology (2017), 1 – 6
Erythropoietin neuroprotection in perinatal asphyxia
RR Malla et al
6
26 Statler PA, McPherson RJ, Bauer LA, Kellert BA, Juul SE. Pharmacokinetics of 32 Ehrenreich H, Weissenborn K, Prange H, Schneider D, Weimar C, Wartenberg K
high-dose recombinant erythropoietin in plasma and brain of neonatal rats. et al. Recombinant human erythropoietin in the treatment of acute ischemic
Pediatr Res 2007; 61: 671–675. stroke. Stroke 2009; 40: e647–e656.
27 McPherson RJ, Demers EJ, Juul SE. Safety of high-dose recombinant 33 Rogers EE, Bonifacio SL, Glass HC, Juul SE, Chang T, Mayock DE et al.
erythropoietin in a neonatal rat model. Neonatology 2007; 91: 36–43. Erythropoietin and hypothermia for hypoxic-ischemic encephalopathy. Pediatr
28 Wu YW, Bauer LA, Ballard RA, Ferriero DM, Glidden DV, Mayock DE et al. Neurol 2014; 51: 657–662.
Erythropoietin for neuroprotection in neonatal encephalopathy: safety and 34 El Shimi MS, Awad HA, Hassanein SM, Gad GI, Imam SS, Shaaban HA et al. Single
pharmacokinetics. Pediatrics 2012; 130: 683–691. dose recombinant erythropoietin versus moderate hypothermia for neonatal
29 Chang YS, Mu D, Wendland M, Sheldon RA, Vexler ZS, McQuillen PS et al. hypoxic ischemic encephalopathy in low resource settings. J Matern Fetal
Erythropoietin improves functional and histological outcome in neonatal stroke. Neonatal Med 2014; 27: 1295–1300.
Pediatr Res 2005; 58: 106–111. 35 Wu YW, Mathur AM, Chang T, McKinstry RC, Mulkey SB, Mayock DE et al.
30 Ehrenreich H, Fischer B, Norra C, Schellenberger F, Stender N, Stiefel M et al. High-dose erythropoietin and hypothermia for hypoxic-ischemic encephalopathy:
Exploring recombinant human erythropoietin in chronic progressive multiple a Phase II Trial. Pediatrics 2016; 137(6): e20160191.
sclerosis. Brain 2007; 130: 2577–2588. 36 Benders MJ, van der Aa NE, Roks M, van Straaten HL, Isgum I,
31 Ehrenreich H, Hinze-Selch D, Stawicki S, Aust C, Knolle-Veentjer S, Wilms S et al. Viergever MA et al. Feasibility and safety of erythropoietin for
Improvement of cognitive functions in chronic schizophrenic patients by neuroprotection after perinatal arterial ischemic stroke. J Pediatr 2014; 164:
recombinant human erythropoietin. Mol Psychiatry 2007; 12: 206–220. 481–486.

Journal of Perinatology (2017), 1 – 6 © 2017 Nature America, Inc., part of Springer Nature.

You might also like