You are on page 1of 8

SE M I N A R S I N P E R I N A T O L O G Y ] (2015) ]]]–]]]

Available online at www.sciencedirect.com

Seminars in Perinatology

www.seminperinat.com

Drugs for neuroprotection after birth asphyxia:


Pharmacologic adjuncts to hypothermia
Frank van Bel, MD, PhDn, and Floris Groenendaal, MD, PhD
Department of Neonatology, University Medical Center Utrecht, Wilhelmina Children’s Hospital, Utrecht,
The Netherlands

article info abstra ct

Keywords: An adverse outcome is still encountered in 45% of full-term neonates with perinatal
Perinatal asphyxia asphyxia who are treated with moderate hypothermia. At present pharmacologic therapies
Pharmacologic neuroprotection are developed to be added to hypothermia. In the present article, these potential neuro-
Hypothermia protective interventions are described based on the molecular pathways set in motion
during fetal hypoxia and following reoxygenation and reperfusion after birth. These
pathways include excessive production of excitotoxins with subsequent over-stimulation
of NMDA receptors and calcium influx in neuronal cells, excessive production of reactive
oxygen and nitrogen species, activation of inflammation leading to inappropriate apopto-
sis, and loss of neurotrophic factors. Possibilities for pharmacologic combination therapy,
where each drug will be administered based on the optimal point of time in the cascade of
destructive molecular reactions, may further reduce brain damage due to perinatal
asphyxia.
& 2015 Elsevier Inc. All rights reserved.

Introduction and necrotic neuronal cell death due to secondary energy


failure. This delayed brain damage may proceed up to days or
Perinatal asphyxia in the near-term and full-term infant is even weeks after birth.3,4
still one of the most important causes of neonatal death and At the same time this biphasic or even triphasic pattern of
adverse motor and cognitive outcome and has an incidence brain damage creates a “therapeutic” window, which is
of 2–20 in every 1000 live born infants, depending in which believed to range from birth up to 6 h of life, during which
part of the world they are born.1,2 neuroprotective strategies can prevent or reduce the full
There is increasing evidence that not only the actual period consequences of delayed brain damage.4–6 Up to now, the
of antenatal and/or perinatal hypoxia-ischemia (HI) affects only established postnatal therapy to achieve this goal to a
the neuronal cells, but also reoxygenation and reperfusion certain extent is moderate hypothermia.7,8 Although the
upon and after birth add substantially to delayed apoptotic composite adverse outcome was reduced, an adverse

Herewith the authors of this article, Frank van Bel, MD and Floris Groenendaal, MD, declare that Frank van Bel and Floris Groenendaal
are, together with Cacha Peeters-Scholte, inventors of 2-iminobiotin as neuroprotective agent for neonates with cerebral hypoxia-
ischemia. They have any financial or personal relationships with other people or organizations that could potentially and
inappropriately influence their work and conclusions. They further declare to have no proprietary or commercial interest in any
product mentioned or concept discussed in this article.
n
Correspondence to: Department of Neonatology, University Medical Center Utrecht, Wilhelmina Children’s Hospital, Room KE 04.123.1,
P.O. Box 85090, 3508 AB Utrecht, The Netherlands.
E-mail address: f.vanbel-2@umcutrecht.nl (F.v. Bel).

http://dx.doi.org/10.1053/j.semperi.2015.12.003
0146-0005/& 2015 Elsevier Inc. All rights reserved.
2 SE M I N A R S I N P E R I N A T O L O G Y ] (2015) ]]]–]]]

Table – Information of dosages of drugs and gasses used or being used in (ongoing) clinical studies investigating
pharmacologic neuroprotection after perinatal hypoxia-ischemia.

Drug or gas Dosage

Maternal allopurinol
Torrance et al.30 500 mg iv; IT: 10 min
Kaandorp et al.26 500 mg iv; IT: 10 min

Postnatal allopurinol
Van Bel et al.62 20 mg/kg/iv o 4 h (IT: 10 min); repeat 20 mg/kg/iv after 12 h
Benders et al.99 20 mg/kg/iv o 4 h (IT: 10 min); repeat 20 mg/kg/iv after 12 h
Gunes et al.63 20 mg/kg/iv o 6 h (IT: 10 min); repeat every 12 h (total 120 mg/kg)

Postnatal 2-iminobiotin (CT: NCT01626924) 0.2 mg/kg/dose iv, o6h, repeat every 4 h up to 6 doses in 20 h

Postnatal rhEPO
Elmahdy et al.82 2500 U/kg/sc o 6 h;repeated daily for 5 days
Zhu et al.83 300 U/kg or 500 U/kg/sc o 48 h; every other day for 2 week
Wu et al.84 (þHT) Postnatal 250, 500, 1000, or 2500 U/kg/iv o 24 h; up to 6 doses every 48 h

Darbepoetin
Baserga et al.88 (þHT) 2 or 10 μg/kg/iv o 12 h; second dose at day 7

Postnatal xenon ventilation


TOBYXe study (þHT) (CT: NCT00934700) 30% Xenon gas for 24 h (start o 6 h)
CoolXeno 3 study(þHT) (CT: NCT02071394) 50% Xenon gas for 18 h (start o 5 h)

Postnatal Topiramate
NeoNATI study (þHT) (CT: NCT1241019) 10 mg/kg/orally-at admission; repeat at days 2 and 3

Postnatal MgSO4
Hemen study (þHT) (CT: NCT02499393) 250 mg/kg/iv o 6 h (IT: 60 min); repeat on days 2 and 3.

CT, www.ClinicalTrials.gov; HT, hypothermia; IT, infusion time.

outcome of 45% is still high8 and it is conceivable that the 2. Upon reoxygenation and reperfusion xanthine will be
outcome can be improved further when moderate hypother- metabolized to uric acid at the expense of excessive
mia will be combined with other neuroprotective strategies. formation of the superoxide free radical (O2 d ), which
In order to achieve this goal it is important to delineate the plays a central role in the further formation of free
potentially harmful cascade of molecular pathways induced radicals and toxic compounds11–13: it reacts with pro-
by fetal hypoxia and upon and after reoxygenation and radicals such as non-protein bound iron to form the very
reperfusion. With this knowledge additional pharmacother- toxic hydroxyl free radical (OH∙).14 It furthermore reacts
apeutical interventions can be considered which may be with nitric oxide (NO∙), because of the HI-increased
capable to inhibit these destructive mechanisms. In this formation of neuronal and later also inducible nitric
article we will particularly focus on those pharmacological oxide synthase (nNOS; iNOS),15,16 to form the toxic
interventions which can be expected to be effective in the peroxynitrite (ONOO).
human infant within the near future. Information of proposed The initially high levels of excitatory neurotransmit-
dosages of the drugs and gasses discussed in this article will be ters and the surge of free radicals, especially upon
provided, when available, in a separate table (Table). and early after reperfusion and reoxygenation con-
tribute to activation of transcription factors such as
nuclear factor kappa B (NFkB) and c-jun N-terminal
Destructive molecular pathways initiated during kinase (JNK) with subsequent activation of an
and after HI inflammatory response with abundant formation of
pro- and anti-inflammatory cytokines, and leading to
production of iNOS setting in motion a pre-apoptotic
1. Fetal hypoxia, an important determinant of perinatal
pathway with further (delayed) brain damage.17–19
asphyxia, sets in motion the excessive production of
3. Finally, a substantial part of the delayed brain damage is
excitatory neurotransmitters which gives rise to the
related to downregulation of the formation of neuro-
activation of N-methyl D-aspartate (NMDA)- and
tropic, maturational, and growth factors inhibiting neu-
voltage-regulated ion channels on the cell membranes
rogenesis and repair.20,21
resulting in a surge of extracellular calcium into neuro-
nal and microglial cells and astrocytes.9,10 This will not
only lead to direct cell damage but also to a drop in the
intra-and intercellular pH leading to production of pro- In the Figure A, the four patterns of above mentioned
radicals liberated from their binding proteins and accu- destructive molecular pathways are schematically depicted
mulation of xanthine. as a function of postnatal age. The proposed pharmacological
SEM I N A R S I N P E R I N A T O L O G Y ] (2015) ]]]–]]] 3

A) Time profile destructive pathways induced by perinatal asphyxia


Superoxide FR (O2-•)
(Pro-)radical formation (i.e.OH•)

pH Inflammation/cytokines / iNOS/Apoptosis
nNOS >NO° > ONOO-
FREE-Iron

Ca 2+

glutamate

Hypo-
Xanthine Modulation trophic factors

30 min 3h 6h 12h 24h days


Fetal
hypoxia Birth
B) Optimal neuroprotective (combination) therapy
Maternal/neonatal allopurinol (xenon/argon)
Hypothermia (72h)
Melatonin/rhEPO Melatonin/rhEPO
Sel-nNOS/iNOS inhib (2-IB)
rhEPO/IGF-1/repair with (M)SCs

Fig – (A) The (post-ictal) time profiles of the potentially destructive molecular pathways are depicted as a function of postnatal
age (hours). During fetal hypoxia there is an increase in excitatory neurotransmitters inducing calcium influx into brain cells;
formation of pro-radicals (free iron) due to lowering of the interstitial pH and accumulation of hypoxanthine. Upon
reoxygenation/reperfusion an early surge of O2 d (white line) and consequently of other free radicals (OH∙) (orange line) and
nitric oxide synthase-induced ONOO (blue line) occurs. This chain of events as well activates the inflammatory pathway with
increased formation of pro- and anti-inflammatory cytokines (purple line) from about 6 h onward with switches on
inappropriate apoptotic activity. Finally the neurotrophic factors are downregulated (yellow line) leading to a diminished
possibility of recovery of affected neurons (O2 d , superoxide free radical; OH∙, hydroxyl free radical; ONOO, peroxynitrite). (B)
Possible combination therapy paradigms with optimal points of time for start and cessation of pharmacological compounds
directed to the different destructive molecular pathways. If recognized, maternal allopurinol can be started during substantial
fetal hypoxia and continued during early life. Free radical and ONOO formation can be reduced by free radical scavenging by
melatonin, rhEPO and 2-IB respectively (start within 6 h of age), whereas anti-inflammatory activity can be challenged by
melatonin and rhEPO up to 48 h of age; reduction of apoptosis and stimulation of production of (neuro)trophic factors can be
achieved with rhEPO, IGF-1 and/or (M)SCs, administered for longer periods (days or weeks) (rhEPO, recombinant human
erythropoietin; 2-IB, 2-iminobiotin; IGF-1, insulin growth factor-1; (M)SCs, (mesenchymal) stem cells). (For interpretation of the
references to color in this figure legend, the reader is referred to the web version of this article.)

interventions as described below will be discussed on basis of oxypurinol readily pass the placenta and are detectable in
the course of the postnatal time pattern of these pathways. umbilical blood in therapeutic concentrations as early as
20 min after maternal oral or intravenous administration.23
Especially in high dosages allopurinol is supposed to be a
Pharmacologic neuroprotective strategies (on top free-iron chelator and free-radical (hydroxyl) scavenger.24,25
of hypothermia) Based on short-term outcome markers, allopurinol has been
shown to be potentially effective, but effects were gender
Antenatal neuroprotective therapy specific: only females showed benefit from antenatal therapy
with allopurinol.26 Sex differences with respect to neuro-
The formation of excitatory neurotransmitters, free (pro-) protective therapy after HI were already suggested in earlier
radicals starts already during the actual hypoxia and accel- reports27–29 A drawback of the study was that in the vast
erates upon birth. In this setting antenatal therapy via the majority of fetuses hypoxia was mild. The long-term results
mother can be considered to be the “first line of defense,” at of this study are currently under investigation. An earlier
least when fetal hypoxia has been recognized. pilot study preceding the randomized trial by the same
The only drug investigated in a randomized blinded clinical investigators in fetuses with more severe hypoxia showed
trial in pregnant women just before delivery and diagnosed the potency of allopurinol as a neuroprotective agent.30
with fetal hypoxia is the xanthine-oxidase inhibitor allopur- A second potentially promising neuroprotectant is the
inol and its active metabolite oxypurinol.22 Allopurinol and noble gas xenon. Xenon is an antagonist of the NMDA
4 SE M I N A R S I N P E R I N A T O L O G Y ] (2015) ]]]–]]]

receptor but has also strong anti-apoptotic properties.31–33 To avoid these latter potential problems other noble gases
And, most important, xenon is considered to be a safe have been tested for neuroprotective properties.52 In partic-
anesthetic which rapidly crosses the placenta.34 This makes ular argon was neuroprotective after neonatal brain hypoxia-
xenon very attractive for its use in mothers undergoing an ischemia.52,53 A study in piglets showed cardiovascular stabil-
emergency cesarean section because of severe fetal hypo- ity of argon administration after hypoxia and during ther-
xia.35 However, unlike for postnatal use (see also below), apeutic hypothermia.54 Blood levels of argon could be
there are to date no studies performed to investigate and measured and were dose dependent. The noble gas argon is
confirm the neuroprotective actions of xenon as an antenatal much cheaper than xenon, and may be a useful alternative
neuroprotective strategy. for xenon.
Ascorbic acid (vitamin C),36,37 tetrahydrobiopterin (BH4),38 Another clinically potential pharmacologic intervention for
phenobarbital,39 N-acetylcysteine,40 and melatonin41 were or the near future may be inhibition of excessive NOS formation.
are proposed to be of value for antenatal neuroprotection, but Whereas non-selective inhibition of NOS is detrimental,
an unfavorable safety profile (N-acetylcysteine), inconclusive probably due to vasoconstriction, selective inhibition of
or disappointing clinical results (vitamin C and phenobarbi- neuronal and inducible NOS appears to be neuroprotec-
tal) or no clinical studies (melatonin) prevent clinical use of tive.55,56 A selective nNOS inhibitor that has been used in
these compounds up to now.42 many experiments is 7-nitroindazole. However, some addi-
Magnesium has been shown to reduce excitotoxicity fol- tional inhibition of eNOS may occur, which is not desirable in
lowing hypoxia-ischemia by inhibition of the NMDA-recep- cerebral hypoxia-ischemia.57 Therefore, compounds have
tor/ion channel complex.43 The mode of action of magnesium been developed that have much higher in vitro specificity
in fetal neuroprotection remains largely unknown, since and potency than available nNOS inhibitors.58 Combination
in vivo concentrations are much lower than those needed of a selective nNOS inhibitor with a selective iNOS may be
for inhibition of the NMDA receptor.44 Although magnesium more effective. 2-Iminobiotin is an in vitro inhibitor of both
administration before 33 weeks of gestation to women with nNOS and iNOS and provides neuroprotection after hypoxia-
high-risk pregnancies have resulted in a reduction of cerebral ischemia in neonatal rat and pig models, although the neuro-
palsy and adverse motor development 45 is in term pregnan- protection may be independent of NOS inhibition in vivo.59–61
cies no definite beneficial effects of maternal magnesium A phase 2 study is ongoing (ClinicalTrials.gov Identifier:
demonstrated.46 NCT01626924).
Allopurinol may have its most optimal indication for
Early postnatal neuroprotective therapy antenatal use in case of proven fetal hypoxia, especially to
inhibit the excessive production of xanthine-oxidase during
As summarized above, the immediate postnatal reoxygena- birth. However, several clinical studies reporting the use of
tion–reperfusion phase leads to further to the activation of intravenous allopurinol in the (early) postnatal period
NMDA- and ion channels with subsequent calcium influx in showed a beneficial effect on short-term cerebral biomarkers
neurons, excessive formation of free radicals, and the nNOS- and even on long-term outcome, especially in moderately
related toxic ONOO. asphyxiated infants and seems to have no serious adverse
Therefore, NMDA/ion channel antagonists, inhibition of effects.62–64
xanthine-oxidase, prevention of nNOS (and subsequently However, in a Cochrane review in 2008 no conclusions
also of iNOS) formation, and chelation of pro-radicals are could be drawn about the efficacy of postnatal neuroprotec-
critical to reduce early reperfusion–reoxygenation damage to tion with allopurinol after perinatal asphyxia since the
the brain. clinical trials reported up to now were underpowered.65 An
In recent studies noble gases, which are NMDA/ion channel international trial aiming to treat asphyxiated newborns on
antagonists, have been used for neuroprotection. Xenon, a the resuscitation table with allopurinol is under considera-
known anesthetic has been tested in several experiments.47–49 tion at present.
Xenon shows a rapid passage of the blood–brain barrier, and Other neuroprotective drugs with potential for future clin-
has no reported negative cardiovascular side effects. Xenon as ical use, particularly in the early postnatal period, are mela-
such did not appear to be effective, but in combination with tonin and N-acetylcysteine (NAC). Melatonin has been
hypothermia neuroprotection by xenon has been demon- thought to have anti-oxidative properties on top of its anti-
strated in several species.42 inflammatory and anti-apoptotic effects.66–68 Experimental
In a feasibility study 14 neonates with perinatal asphyxia studies showed neuroprotective properties in the mid- and
and therapeutic hypothermia received up to 50% xenon for late-gestation sheep fetus.68–69 A small clinical study in
18 h and no adverse side effects were demonstrated.50 asphyxiated newborns showed a reduction of free radical
Several dosing strategies of xenon are explored, including formation.70 Although melatonin seems to be safe and with-
50% xenon for 18 h (ClinicalTrials: NCT02071394), or 30% out serious adverse effects, there is no consensus on the
xenon for 24 h (ClinicalTrials: NCT00934700). At present optimal dosing.71 NAC is a precursor of glutathione, has the
follow-up of the patients included in these studies is ongoing. properties of an anti-oxidant and is supposed to exert also a
Prolonged use of xenon in neonates faces several chal- direct free radical scavenging activity.72 The neuroprotective
lenges. Most important of all xenon is very expensive, and properties are controversial and (hemodynamic) adverse
therefore adjustments in neonatal ventilators need to be reactions are reported.42 Neuroprotective actions of NAC have
made to recruit xenon.51 To reduce loss of xenon cuffed tubes been reported in neonatal rodents as well as in piglets with
are used which might enhance the risk of tracheal lesions. and without additional hypothermia.73,74 Although clinically
SEM I N A R S I N P E R I N A T O L O G Y ] (2015) ]]]–]]] 5

used in preterm newborns, mostly in relation with preven- newborns. However, especially when higher dosages are used
tion of bronchopulmonary dysplasia, no clinical studies are and/or administered for longer periods of time there may be
done yet in asphyxiated (term) newborns. concerns with respect to safety and more research is war-
Studies with tetrahydrobiopterin being a neuroprotective ranted here.85,86 EPO derivatives which have less unwanted
agent after perinatal asphyxia are actually non-existent, effects such as asialo-EPO 87 or longer half-life such as
although its safety profile is very attractive for further study darbepoietin88 are less well investigated but may be clinically
in relation with perinatal hypoxia-ischemia.42 Topiramate, a interesting because of their specific properties.
well-known anticonvulsant, has sparsely investigated for its In addition to its anti-oxidative and free radical scavenging
neuroprotective actions after birth asphyxia.75 A safety and actions melatonin is also supposed to have anti-
efficacy study (NeoNATI) was recently completed in combi- inflammatory properties by preventing activation and trans-
nation with moderate hypothermia76 (ClinicalTrials: location of NFkB to the nucleus89 (also see above).
NCT01241019). Postnatal magnesium administration has not The development of inhibitors of pro-inflammatory cyto-
been uniform in demonstrating neuroprotective effects of kines90 or anti-apoptotic strategies (NFkB inhibition)91 are
magnesium in full-term affected neonates.44 Earlier trials for extremely interesting and potentially very forceful but
neuroprotection after perinatal hypoxia-ischemia were dis- beyond the focus of this review.
continued because Magnesium administration carried the
risk of severe hypotension.77 However, currently studies are
Suppletion of (neuro)trophic and growth factors
performed of magnesium combined with moderate hypo-
thermia (ClinicalTrials: NCT02499393 and NCT01646619).
Neurotrophic, maturational, and growth factors are impor-
tant for a normal development of the immature brain by
Anti-inflammatory and anti-apoptotic therapy
stimulation of the endogenous neurogenesis.92 Downregula-
tion of these factors occurs after severe perinatal HI and may
The transcription factor nuclear factor kappa B (NFkB) regu-
be deleterious for recovery and proper development of the
lates the expression of genes involved in inflammation and
post-asphyxial newborn brain.
apoptotic activity. Its activation gives rise to abundant for-
Besides the anti-oxidative and anti-inflammatory actions of
mation of pro- and anti-inflammatory cytokines, production
EPO, as described above, it has been supposed that EPO has
of inducible NOS (iNOS), and inappropriate apoptosis.18
also important neurotrophic properties.93–95
Of the neuroprotective agents with an anti-inflammatory
It is suggested that for a sustained effect of EPO on neuro-
action and a potential for clinical application in the near
and angiogenesis and thus improved long-term outcome
future erythropoietin (EPO) is the most and up to now only
higher doses (up to 5000 U/kg) and prolonged periods of
attractive candidate.78 EPO exerts its anti-inflammatory
treatment are necessary, which may raise safety
action after binding to its receptor (EPOR) which is expressed
concerns.85,86
on the membranes of neurons, astrocytes, and microglial
An important related issue is the role of hypoxia-induced
cells.79 Recombinant Human EPO (rhEPO) is already quite
factor 1α (HIF-1α), activated during and upon hypoxia-ische-
extensively used in the very and extremely preterm infant in
mia, which gives rise to the formation of transcriptional
increasing dosages and in a pilot study in term neonates with
targets, leading to an enhanced production of EPO and
neonatal stroke and has a safe pharmacological profile.80,81
endothelial growth factor and consequent brain repair.96
Numerous experimental studies in experimental studies in
Other neurotrophic factors such as IGF-1, basic fibroblast
different neonatal species showed its neuroprotective proper-
growth factor (bFGF), and BDNF in experimental studies in
ties with respect to post-hypoxic-ischemic brain damage.79
neonatal animals are reported to be also effective to reduce
Clinically a pilot study of 45 patients showed that treatment
post-asphyxial brain damage.97,98 However, use of autologous
of asphyxiated newborns with rhEPO seemed safe and fea-
or allogeneic mesenchymal stem cell therapy after perinatal
sible.82 A blinded randomized phase III trial in asphyxiated
hypoxia-ischemia seems more effective in this respect but is
term newborns in China showed that repeated (low) doses of
discussed separately in this issue.99
intravenous administered rhEPO decreased the incidence of
disabilities at 18 months of age, although there was a
suggestion that females but not necessarily males benefit Combination therapy, the way to go
from this therapy.83 A pharmacokinetic study in 24 severely
asphyxiated newborns who underwent hypothermia were It is unrealistic to assume that single or add-on therapy
treated with additional rhEPO and showed that a quite high without taking into account the time frame of the sequential
dose of 1000 U/kg was well tolerated. Plasma concentrations potentially destructive molecular pathways, will be the ulti-
were similar as reported in earlier neuroprotective studies in mate answer toward a substantial reduction of brain damage
newborn animals.84 A randomized trial with hypothermia after (severe) perinatal asphyxia. Moreover, appropriate dos-
with high-dose rhEPO as add-on therapy is currently running ing of the drugs, pharmacologic compounds, and gasses
(ClinicalTrials: NCT01913340) These randomized trials in discussed above, the duration and site of their administra-
newborns undergoing hypothermia are necessary to ulti- tion, and a possible gender specificity of the compounds used
mately confirm the neuroprotective actions of additional needs ongoing research. However, when we can solve the
rhEPO and to determine the optimal timing, dose, and above mentioned questions and are able to determine the
duration of treatment. Up to now no serious adverse effects optimal post-ictal time of intervention in each specific
are reported during the clinical use of rhEPO in (preterm) molecular pathway, a major achievement can be added to
6 SE M I N A R S I N P E R I N A T O L O G Y ] (2015) ]]]–]]]

the treatment of hypoxia-ischemia-induced neonatal 13. Mink R, Johnston J. The effect of infusing hypoxanthine or
encephalopathy.78 xanthine on hypoxic-ischemic brain injury in rabbits. Brain
The Figure B shows a tentative (pharmacological) treatment Res. 2007;1147:256–264.
14. Shouman BO, Mesbah A, Aly H. Iron metabolism and lipid
paradigm, which may have clinical relevance in the (near)
peroxidation products in infants with hypoxic ischemic
future.
encephalopathy. J Perinatol. 2008;28:487–491.
15. Van den Tweel ER, Nijboer C, Kavelaars A, Heijnen CJ,
Groenendaal F, van Bel F. Expression of nitric oxide synthase
isoforms and nitrotyrosine formation after hypoxia-ischemia
in the neonatal rat brain. J Neuroimmunol. 2005;167:64–71.
Concluding remarks 16. Suzuki M, Tabuchi M, Ikeda M, Tomita T. Concurrent for-
mation of peroxynitrite with the expression of inducible nitric
Ongoing research in neuroprotective pharmacology after oxide synthase in the brain during middle cerebral artery
perinatal asphyxia has offered us potentially forceful com- occlusion and reperfusion in rats. Brain Res. 2002;951:113–120.
pounds to further reduce and to repair damage to the 17. Leonardo CC, Pennypacker KR. Neuroinflammation and
immature brain during and after severe birth asphyxia as MMPs: potential therapeutic targets in neonatal hypoxic-
ischemic injury. J Neuroinflamm. 2009;6:13.
add-ons to the only established neuroprotective therapy—
18. Nijboer CH, Heijnen CJ, Groenendaal F, May MJ, van Bel F,
moderate hypothermia. There is still a long way to go to
Kavelaars A. A dual role of the NF-kappa B pathway in
determine the optimal dose of these compounds and to find neonatal hypoxic-ischemic brain damage. Stroke. 2008;39:
the optimal point of time of start and duration of pharmaco- 2578–2586.
logical therapy. A definite decision on the clinical use 19. Hedtjarn M, Mallard C, Hagberg H. Inflammatory gene profil-
depends, of course, first of all on the safety profile of the ing in the developing mouse brain after hypoxia-ischemia. J
potentially neuroprotective drug. Cereb Blood Flow Metab. 2004;24:1333–1351.
20. Scheepens A, Wassink G, blanco CE. The effect of global birth
r e f e r e n c e s asphyxia on the ontogeny of BDNF and NGF protein expression
in the juvenile brain. Brain Res Dev Brain Res. 2003;140:215–221.
21. Skoff RP, Bessert D, Barks JD, Silverstein FS. Plasticity of
neurons and glia following neonatal hypoxic-ischemic brain
1. Glass HC, Ferriero DM. Treatment of hypoxic-ischemic ence-
injury in rats. Neurochem Res. 2007;32:331–342.
phalopathy in newborns. Curr Treat Options Neurol. 2007;9:
22. Boda D. Results of and further prevention of hypoxic fetal
414–423.
brain damage by inhibition of xanthine oxidase enzyme with
2. Aminu M, Unkels R, Mdegela m, Utz B, Adaji S, van den Broek
allopurinol. J Perinat Med. 2011;39:441–444.
N. Causes of and factors associated with stillbirth in low- and
23. Boda D, Nemeth P, Kiss P, et al. Treatment of mothers with
middle-income countries: a systematic literature review. Int J
allopurinol to produce therapeutic blood levels in newborns.
Obstet Gynaecol. 2014;121(suppl 4):141–453.
Prenat Neonat Med. 1999;4:130–132.
3. Lorek A, Takei Y, Cady E, et al. Delayed (“secondary”) cerebral
24. Ko KM, Godin DV. Inhibition of transition metal ion-catalysed
energy failure after acute hypoxia-ischemia in the newborn
ascorbate oxidation and lipid peroxidation by allopurinol and
piglet: continuous 48-hour studies by phosphorus magnetic
oxypurinol. Biochem Pharmacol. 1990;40:803–809.
resonance spectroscopy. Pediatr Res. 1994;36:669–676.
4. Iwata O, Iwata S, Bainbridge A, et al. Supra- and sub-baseline 25. Moorhouse PC, Grootveld M, Halliwell B, Quilan JG, Gutteridge
phosphocreatine recovery in developing brain after transient JM. Allopurinol and oxypurinol are hydroxyl radical scav-
hypoxia-ischaemia: relation to baseline energetics, insult engers. FEBS Lett. 1987;213:23–28.
severity and outcome. Brain. 2008;131:2220–2226. 26. Kaandorp JJ, Benders JNL, Schuit E, et al. Maternal allopurinol
5. Hope PL, Costello AM, Cady EB, et al. Cerebral energy metab- administration during suspected fetal hypoxia: a novel neu-
olism studied with phosphorous NMR spectroscopy in normal roprotective intervention? A multicentre randomized placebo
and birth asphyxiated infants. Lancet. 1984;8399:366–370. controlled trial. Arch Dis Child Fetal Ed. 2014;100:216–223.
6. Fleiss B, Gressens P. Tertiary mechanisms of brain damage: a 27. Nijboer CH, Kavelaars A, van Bel F, Heijnen CJ, Groenendaal F.
new hope for treatment of cerebral palsy? Lancet Neurol. Gender-dependent pathways of hypoxia-ischemia-induced
2012;11:556–566. cell death and neuroprotection in the immature P3 rat. Dev
7. Thoresen M, Whitelaw A. Therapeutic hypothermia for Neurosci. 2007;29:385–392.
hypoxic-ischaemic encephalopathy in the newborn infant. 28. Weis SN, Pettenuzzo LF, Krolow R, et al. Neonatal hypoxia-
Curr Opin Neurol. 2005;18:111–116. ischemia induces sex-related changes in rat brain mitochon-
8. Jacobs SE, Berg M, Tarnow-Modi WO. Cooling for newborns dria. Mitochondrion. 2012;12:271–279.
with hypoxic ischaemic encephalopathy. Cochrane Database 29. Hill CA, Fitch RH. Sex differences in mechanisms and out-
Syst Rev. 2013;1:CD003311. come of neonatal hypoxia-ischemia in rodent models: impli-
9. Ferriero DM. Neonatal brain injury. N Engl J Med. 2004;351: cations for sex-specific neuroprotection in clinical neonatal
1985–1995. practice. Neurol Res Internat. 2012;2012:1–9.
10. Hilton GD, Nunez JL, Bambrick L, Thompson SM, McCarthy 30. Torrance HL, Benders MJ, Derks JB, et al. Maternal allopurinol
MM. Glutamate-mediated excitotoxicity in neonatal hippo- during fetal hypoxia lowers cord blood levels of brain injury
campal neurons is mediated by mGlur-induced release of marker S-100B. Pediatrics. 2009;124:350–357.
Caþþ from intracellular stores and is prevented by estradiol. 31. Franks N, Dickinson R, de Sousa S, Hall A, Lieb W. How does
Eur J Neurosci. 2006;24:3008–3016. Xenon produce anaesthesia? Nature. 1998;26:324.
11. Saugstad OD. Role of xanthine oxidase and its inhibitor 32. Ma D, Williamson P, Januszewski A, et al. Xenon mitigates
in hypoxia: reoxygenation injury. Pediatrics. 1996;98:103–107. isoflurane-induced neuronal apoptosis in the developing
12. Ono T, Tsuruta R, Fujita M, et al. Xanthine oxidase is one of rodent brain. Anaesthesiology. 2007;106:746–753.
the major sources of superoxide anion radicals in blood after 33. Ma D, Lim T, Xu J, et al. Xenon preconditioning protects
reperfusion in rats with forebrain ischemia/reperfusion. Brain against renal ischemic-reperfusion injury via HIF-1alpha
Res. 2009;1305:158–167. activation. J Am Soc Nephrol. 2009;20:713–720.
SEM I N A R S I N P E R I N A T O L O G Y ] (2015) ]]]–]]] 7

34. Goto T, Saito H, Shinkai M, Nakata Y, Ichinose F, Morita S. 53. Ryang YM, Fahlenkamp AV, Rossaint R, et al. Neuroprotective
Xenon provides faster emergence from anesthesia than does effects of argon in an in vivo model of transient middle cerebral
nitrous oxide sevoflurane or nitrous-isoflurane. Anaesthesiol- artery occlusion in rats. Crit Care Med. 2011;39:1448–1453.
ogy. 1997;86:1273–1278. 54. Alderliesten T, Favie LM, Neijzen RW, et al. Neuroprotection
35. Lane GA, Nahrwold ML, Tait AR, Taylor-Busch M, Cohen PJ, by argon ventilation after perinatal asphyxia: a safety study in
Beaudoin AR. Anesthetics as teratogens: nitrous oxide is newborn piglets. PLoS One. 2014;9:e113575.
fetotoxic, xenon is not. Science. 1980;210:899–901. 55. van den Tweel ERW, Peeters-Scholte CMPCD, Van Bel F,
36. Jackson T, Xu A, Vita J, Keaney JJ. Ascorbate prevents the Heijnen CJ, Groenendaal F. Inhibition of nNOS and iNOS
interaction of superoxide and nitric oxide only at very high following hypoxia-ischemia improves long-term outcome
physiological concentrations. Circ Res. 1998;83:916–922. but does not influence the inflammatory response in the
37. Poston L, Briley AL, Seed PT, Kelly FJ, Shennan AH. Vitamins in neonatal rat brain. Dev Neurosci. 2002;24:389–395.
pre-eclampsia (VIP) trial consortium. Vitamin C and vitamin E 56. Willmot M, Gibson C, Gray L, Murphy S, Bath P. Nitric oxide
in pregnant women at risk for pre-eclampsia (VIP trial): random- synthase inhibitors in experimental ischemic stroke and their
ized placebo-controlled trial. Lancet. 2006;367:1145–1154. effects on infarct size and cerebral blood flow: a systematic
38. Vasquez-Vivar J, Whitsett J, Derrick M, Ji X, Yu L, Tan S. review. Free Radic Biol Med. 2005;39:412–425.
Tetrahydrobioptin in the prevention of hypertonia in hypoxic 57. Nakata M, Anno K, Sugino N, Matsumori LT, Nakamura Y,
fetal brain. Ann Neurol. 2009;66:323–331. Kato H. Effect of intrauterine ischemia-hypoxia on endothe-
39. Reinisch JM, Sanders SA, Mortensen EL, Rubin DB. In utero lial nitric oxide synthase in fetal brain in rats. Biol Neonate.
exposure to phenobarbital and intelligence deficits in adult 2002;82:117–121.
men. J Am Med Assoc. 1995;274:1518–1525. 58. Ji H, Tan S, Igarashi J, et al. Selective neuronal nitric oxide
40. Xu D, Chen Y, Wang H, Zhao L, Wang J, Wei W. Effect of synthase inhibitors and the prevention of cerebral palsy. Ann
N-acetylcysteine on lipopolysaccharide-induced intra-uterine Neurol. 2009;65:209–217.
fetal death and intra-uterine growth retardation in mice. 59. Nijboer CH, Groenendaal F, Kavelaars A, Hagberg HH, Van Bel
Toxicol Sci. 2005;88:525–533. F, Heijnen CJ. Gender-specific neuroprotection by 2-
41. Jaynke G, Marr M, Myeris C, Wilson R, Travlos G, Price C. iminobiotin after hypoxia-ischemia in the neonatal rat via a
Maternal and developmental toxicity evaluation of melatonin nitric oxide independent pathway. J Cereb Blood Flow Metab.
administered orally to pregnant Sprague-Dawley rats. Toxicol 2007;27:282–292.
Sci. 1999;50:271–279. 60. Peeters-Scholte C, Koster JG, Veldhuis W, et al. Neuroprotection
42. Robertson NJ, Tan S, Groenendaal F, et al. Which neuro- by selective nitric oxide synthase inhibition at 24 hours after
portective agents are ready for bench to bedside translation perinatal hypoxia-ischemia. Stroke. 2002;33:2304–2310.
in the newborn infant? J Pediatr. 2012;160:544–552. 61. Bjorkman ST, Ireland Z, Fan X, et al. Short-term dose-
43. Lipton SA, Rosenberg PA. Excitatory amino acids as a final response characteristics of 2-iminobiotin immediately post-
common pathway for neurologic disorders. N Engl J Med. insult in the neonatal piglet after hypoxia-ischemia. Stroke.
1994;330:613–622. 2013;44(3):809–811.
44. Galinsky R, Bennet L, Groenendaal F, et al. Magnesium is not 62. Van Bel F, Shadid M, Moison RM, et al. Effect of allopurinol on
consistently neuroprotective for perinatal hypoxia-ischemia postasphyxial free radical formation, cerebral hemodynam-
in term-equivalent models in preclinical studies: a systematic ics, and electrical brain activity. Pediatrics. 1998;101:185–193.
review. Dev Neurosci. 2014;36:73–82. 63. Gunes T, Ozturk MA, Koklu E, Kose K, Gunes I. Effect of
45. Crowther CA, Brown J, McKinlay CJ, Middleton P. Magnesium allopurinol supplementation on nitric oxide levels in asphyxi-
sulphate for preventing preterm birth in threatened preterm ated newborns. Pediatr Neurol. 2007;36:17–24.
labour. Cochrane Database Syst Rev. 2014;8:CD001060. http://dx. 64. Kaandorp J, van Bel F, Veen S, et al. Long-term neuroprotec-
doi.org/10.1002/14651858.CD001060.pub2. tive effects of allopurinol after moderate perinatal asphyxia.
46. Nguyen TM, Crowther CA, Wilkinson D, Bain E. Magnesium Follow-up of two randomized controlled trials. Arch Dis Child.
sulphate for women at term for neuroprotection of the fetus. 2012;97:F162-6.
Cochrane Database Syst Rev. 2013;2:CD009395. http://dx.doi.org/ 65. Chaudhari T, McGuire W. Allopurinol for preventing mortality
10.1002/14651858.CD009395.pub2. and morbidity in newborn infants with suspected hypoxic-
47. Thoresen M, Hobbs CE, Wood T, Chakkarapani E, Dingley J. ishaemic encephalopathy. Cochrane Database Syst Rev. 2008;2:
Cooling combined with immediate or delayed xenon inhala- CD006817.
tion provides equivalent long-term neuroprotection after 66. Boutin JA, Audinot V, Ferry G, delagrange P. Molecular tools to
neonatal hypoxia-ischemia. J Cereb Blood Flow Metab. study melatonin pathway and actions. Trends Pharmacol Sci.
2009;29:707–714. 2005;26:412–419.
48. Faulkner S, Bainbridge A, Kato T, et al. Xenon augmented 67. Hardeland R. Antioxidative protection by melatonin: multi-
hypothermia reduces early lactate/NAA and cell death in plicity of mechanisms from radical detoxification to radical
perinatal asphyxia. Ann Neurol. 2011;70:133–150. avoidance. Endocrine. 2005;27:119–130.
49. Liu X, Dingley J, Scull-Brown E, Thoresen M. Adding 5 h 68. Welin AK, Svedin P, Lapatto R, et al. Melatonin reduces
delayed xenon to delayed hypothermia treatment improves inflammation and cell death in white matter in the mid-
long-term function in neonatal rats surviving to adulthood. gestation fetal sheep following umbilical cord occlusion.
Pediatr Res. 2015;77:779–783. Pediatr Res. 2007;61:153–158.
50. Dingley J, Tooley J, Liu X, et al. Xenon ventilation during 69. Miller SL, Yan EB, Castillo-Melendez M, Jenkin G, Walker DW.
therapeutic hypothermia in neonatal encephalopathy: a feasibility Melatonin provides neuroprotection in the late-gestation
study. Pediatrics, 133; 809–818. fetal sheep brain in reponse to umbilical cord occlusion. Dev
51. Chakkarapani E, Thoresen M, Hobbs CE, Aquilina K, Liu X, Neurosci. 2005;27:200–210.
Dingley J. A closed-circuit neonatal xenon delivery system: a 70. Fulia F, Gitto E, Cuzzocrea S, et al. Increased levels of malon-
technical and practical neuroprotection feasibility study in dialdehyde and nitrite/nitrate in the blood of asphyxiated
newborn pigs. Anesth Analg. 2009;109:451–460. newborns: reduction by melatonin. J Pineal Res. 2005;31:343–349.
52. Zhuang L, Yang T, Zhao H, et al. The protective profile of 71. Robertson N, Faulkner S, Fleiss B, et al. Melatonin augments
argon, helium, and xenon in a model of neonatal asphyxia in hypothermic neuroprotection in a perinatal asphyxia model.
rats. Crit Care Med. 2012;40:1724–1730. Brain. 2013;136:90–105.
8 SE M I N A R S I N P E R I N A T O L O G Y ] (2015) ]]]–]]]

72. Aruoma O, Halliwell B, Hoey B, Butler J. The antioxidant 87. Siren AL, Fasshauer T, Bartels C, Ehrenreich H. Therapeutic
action of N-acetylcysteine: its reaction with hydrogennper- potential of erythropoietin and its structural or functional variants
oxide; hydroxyl radical, superoxide, and hypochlorous acid. in the nervous system. Neurotherapeutics. 2009;6:108–127.
Free Radic Biol Med. 1989;6:593–597. 88. Baserga MC, Beachy JC, Roberts JK, et al. Darbepoetin admin-
73. Plaisant F, Clippe A, VanDer Stricht D, Knoops B, Gressens P. istration to neonates undergoing cooling for encephalopathy
Recombinant peroxiredoxin 5 protects against excitotoxic (DANCE): a safety and pharmacokinetic trial. Pediatr Res.
brain lesions in newborn mice. Free Radic Biol Med. 2003;34: 2015;78:315–322.
862–872. 89. Reiter RJ, Calvo JR, Karbownik M, Qi W, Tan DX. Melatonin
74. Jatana M, Singh I, Singh A, Jenkins D. Combination of and its relation to the immune system and inflammation.
systemic hypothermia and N-acetylcysteine attenuates Ann NY Acad Sci. 2000;917:376–386.
hypoxic-ischemic brain injury in neonatal rats. Pediatr Res. 90. Nijboer CH, Heijnen CJ, Groenendaal F, van Bel F, Kavelaars A.
2006;59:684–689. Alternate pathways preserve tumor necrosis factor-alpha
75. Schubert S, Brandl U, Brodhun M, et al. Neuroprotective production after nuclear factor-kappaB inhibition in neonatal
effects of topiramate after hypoxia-ischemia in newborn cerebral hypoxia-ischemia. Stroke. 2009;40:3362–3368.
piglets. Brain Res. 2005;1058:129–136. 91. Nijboer CH, Heijnen CJ, Groenendaal F, May MJ, van Bel F,
76. Filippi L, Fiorini P, Daniotti M, et al. Safety and efficacy of Kavelaars A. Strong neuroprotection by inhibition of NF-
topiramate in neonates with hypoxic-ischemic encephalop- kappaB after neonatal hypoxia-ischemia involves apoptotic
athy treated with hypothermia (NeoNATI). BMC Pediatr. mechanisms but is independent of cytokines. Stroke.
2012;12:144. 2008;39:2129–2137.
77. Levene M, blennow M, Whitelaw A, et al. Acute effects of two 92. Alagappan D, Lazzarino DA, Felling RJ, et al. Brain injury
different doses of magnesium sulphate in infants with birth expands the numbers of neural stem cells and progenitors in
asphyxia. Arch Dis Child Fetal Neonatal Ed. 1995;73:F174–F177. the SVZ by enhancing their responsiveness to EGF. ASN Neuro.
78. Fan X, Kavelaars A, Heijnen CJ, Groenendaal F, Van Bel F. 2009;1:e00009.
Pharmacological neuroprotection after perinatal hypoxic- 93. Campana WM, Misasi R, O’Brien JS. Identification of a neuro-
ischemic brain injury. Cur Neuropharmacol. 2010;8:324–334. trophic sequence in erythropoietin. Int J Mol Med. 1998;1:
79. Van der Kooij MA, Groenendaal F, Kavelaars A, Heijnen CJ, 235–241.
van Bel F. Neuroprotective properties and mechanisms of 94. Iwai M, Stetler RA, Xing J, et al. Enhanced oligodendrogenesis and
erythropoietin in in vitro and in vivo experimental models for recovery of neurological function by erythropoietin after neonatal
hpoxia/ischemia. Brain Res Rev. 2008;59:22–33. hypoxic/ischemic brain injury. Stroke. 2010;41:1032–1037.
80. Fauchere JC, Koller BM, Tschopp A, et al. Safety of early high- 95. Reitmeir R, Kilic E, Kilic U, et al. Post-acute delivery of
dose recombinant erythropoietin for neuroprotection in very erythropoietin induces stroke recovery by promoting perile-
preterm infants. J Pediatr. 2015;167:52–57. sional tissue remodelling and contralesional pyramidal tract
81. Benders MJ, van der Aa NE, Roks M, et al. Feasibility and plasticity. Brain. 2011;134:84–99.
safety of erythropoietin for neuroprotection after perinatal 96. Fan X, Heijnen CJ, van der Kooij MA, Groenendaal F, van Bel F.
arterial ischemic stroke. J Pediatr. 2014;164:481–486. The role and regulation of hypoxia-inducible factor 1 alpha
82. Elmahdy H, El-Mashad A, El-Bahrawy H, El-Gohary T, expression in brain development and neonatal hypoxic-
El-Barbary A, Aly H. Human recombinant erythropoietin in ischemic brain injury. Brain Res. 2009;62:99–108.
asphyxia neonatorum: pilot trial. Pediatrics. 2010;125:e1135–e1142. 97. Lin S, Fan LW, Rhodes PG, Cai Z. Intranasal administration of
83. Zhu C, Kang W, Xu F, et al. Erythropoietin improved neuro- IGF-1 attenuates hypoxic-ischemic brain injury in neonatal
logic outcomes in newborns with hypoxic-ischemic ence- rats. Exp Neurol. 2009;217:361–370.
phalopathy. Pediatrics. 2009;124:e218–e226. 98. Galvin KA, Oorschot DE. Continuous low-dose treatment with
84. Wu Y, Bauer LA, Ballard RA, et al. Erythropoietin for neuro- brain-derived neurotrophic factor or neurotrophin-3 protects
protecction in neonatal encephalopathy: safety and pharma- striatal medium spiny neurons from mild neonatal hypoxia/
cokinetics. Pediatrics. 2012;130:683–691. ischemia: a stereological study. Neuroscience. 2003;118:
85. Cilio MR, Ferriero DM. Synergic neuroprotective therapies 1023–1032.
with hypothermia. Semin Fetal Neonatal Med. 2010;15:293–298. 99. Benders MJ, Bos AF, Rademaker CM, et al. Early postnatal
86. Eherenreich H, Weissenborn K, Prange H, et al. Recombinant allopurinol does not improve short term outcome after severe
human erythropoietin in the treatment of acute ischemic birth asphyxia. Arch Dis Child Fetal Neonatal Ed. 2006;91:
stroke. Stroke. 2009;40:e647–e656. F163–F165.

You might also like