Você está na página 1de 8

Seminars in Fetal & Neonatal Medicine (2006) 11, 260e267

a v a i l a b l e a t w w w. s c i e n c e d i r e c t . c o m

j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / s i n y

Pain control: Opioid dosing, population kinetics and


side-effects
Sinno H.P. Simons a,b,*, K.J.S. Anand c,d

a
Department of Pediatric Surgery, Erasmus-MC/Sophia Children’s Hospital, Rotterdam, The Netherlands
b
Department of Neonatology, Campus Virchow Klinikum, Charité, Humboldt University Medical Center, Berlin, Germany
c
University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas, USA
d
Pain Neurobiology Laboratory, Arkansas Children’s Hospital Research Institute, Arkansas Children’s Hospital,
Little Rock, USA

KEYWORDS Summary Neonates undergoing invasive procedures, postoperative pain or ventilatory sup-
Fentanyl; port commonly receive opioids for treating pain and stress. Randomized clinical trials have
Infant-newborn; examined the benefits and adverse effects of morphine or fentanyl for ventilated neonates
Morphine; and other indications. This paper summarizes the current evidence for opioid dosing in new-
Pain; borns, reviews their side-effects and explains the use of population kinetics and non-linear
Stress; mixed-effects modeling to analyze the data from clinical trials. Opioid use should be reserved
Surgery for severe pain postoperatively or during intensive care in neonates, using continuous infusions
rather than intermittent boluses. The safety and efficacy data from prolonged opioid use, par-
ticularly on the long-term outcomes of neonates, is still lacking. The pharmacodynamics and
pharmacogenetics of opioid use in infancy needs further investigation, using non-linear
mixed-effects models to drive individualized therapy. The current interest in opioid research
will reap rich dividends in providing pain relief for neonates and avoiding dangerous side effects.
ª 2006 Published by Elsevier Ltd.

Introduction distribution, metabolism and elimination, and variable


severity of illnesses, together with rapid changes in brain
development, receptor expression, opioid pharmacody-
Opioids form the mainstay for treating moderate or severe
namics, and pain mechanisms during the third trimester
pain in preterm and term newborn infants. Opioid dosing
of gestation and early infancy.
is complicated in this patient population because of
Research during the last two decades has focused on
major differences in body composition, drug absorption,
opioid pharmacokinetics/pharmacodynamics (PK/PD) in
clinical trials and methods for pain assessment in the
* Corresponding author. Department of Pediatric Surgery,
newborn infant, but much greater evidence is needed to
Erasmus-MC/Sophia Children’s Hospital, Dr Molewaterplein 60, develop a scientific rationale for the optimal use of
3015 GJ, Rotterdam, The Netherlands. Tel.: C31 1 0244 9923/ analgesic agents in neonates. Clinical analgesic trials in
C49 3 0447 14462; fax: C31 1 0463 6288. newborns are limited by the lack of gold standards to assess
E-mail address: sinnosimons@hotmail.com (S.H.P. Simons). the different types of pain (injury-related, neuropathic,

1744-165X/$ - see front matter ª 2006 Published by Elsevier Ltd.


doi:10.1016/j.siny.2006.02.008
Pain control 261

visceral, inflammatory) and their combinations in different


Table 1 When to consider using opioids in neonates
clinical settings (postoperative pain, burn pain, birth
trauma, critical illness). Objective methods for the assess- Painful procedures Ongoing or frequent
ment of neonatal pain have focused on acute, tissue-injury pain from
related pain, using constructs for only the intensity of pain. Peripheral arterial Mechanical ventilation
Furthermore, ethical problems restrain the performance of or venous cutdown and suctioning
well-designed trials. For instance, the number of blood Central venous Birth trauma
samples that can be taken for opioid plasma level line placement
measurement is limited, and these cannot always be taken Peripherally inserted Indwelling chest
at predetermined times because of the different needs for central catheter placement tubes or other catheters
clinical blood sampling. To support clinical decision making Endotracheal intubationa Meningitis
with reasonable costebenefit analyses for opioid use in Chest tube insertion Necrotizing enterocolitis
neonates, a clearer definition of unwanted opioid side Circumcision Thermal or chemical burns
effects and adverse effects, together with their dose- Other ongoing
related incidence and their incidence in vulnerable severe pain states
populations of neonates, is required. Some of these areas a
In combination with atropine and non-depolarizing muscle
are discussed in this paper, to better inform the clinician at relaxant (pancuronium, vercuronium, rorcuronium).
the bedside of a seriously ill newborn infant.
patients. Adequate analgesic therapy requires clinically
Opioid dosing driven titration towards the individual infant’s needs.
The optimal use of opioids for neonatal patients during
Opioid therapy is generally reserved for clinical situations mechanical ventilation is still under discussion. Opioid
associated with severe pain. For neonatal patients, opioids infusions seem more beneficial than intravenous midazolam
are mostly used during surgical and non-surgical proce- for sedation in ventilated neonates, as concluded in a re-
dures, for postoperative pain and intensive care.1e3 Novel cently performed meta-analysis by the Pain Control Group of
study designs will be required to clarify the dosing sched- the NICHD/FDA Newborn Drug Development Initiative.2 Lim-
ules for commonly used opioids, like morphine or fentanyl, ited evidence is available from studies comparing the bene-
in different clinical situations.4 ficial or side effects of fentanyl and morphine.9,10,27,28
Fentanyl is a synthetic opioid with activity on m1- and Fentanyl has greater analgesic potency, a faster onset and
d-opioid receptors, used frequently in neonates because shorter duration of action than morphine. Fentanyl analgesia
of rapid analgesia,5 hemodynamic stability, blocking stress might be preferred for short periods of time because toler-
responses,6,7 and preventing increases in pulmonary vascu- ance develops more rapidly to fentanyl than to morphine,10
lar resistance.8 Adult studies suggested that fentanyl is whereas morphine might be preferred for prolonged use.
80e100 times more potent than morphine but neonatal Saarenmaa et al. compared both agents in a randomized trial
studies have estimated a potency ratio of 13e20.9 Fentanyl and showed less frequent gastrointestinal dysmotility in fen-
is highly lipophilic, crosses the blood brainebarrier rapidly, tanyl-treated newborns,9 although this study did not include
accumulates in fatty tissues, and causes less histamine a placebo control group. Clinical trials comparing fentanyl
release than morphine. Tolerance to fentanyl develops with placebo are limited but show reduced pain/stress in
more rapidly than to morphine,10 requiring the escalation ventilated newborns after a single dose of fentanyl,29 as
of doses during prolonged administration.11,12 well as continuous fentanyl infusions.6,30
Morphine produces analgesia via activity on m1- and m2- Morphine has been investigated more frequently and
opioid receptors, reduces behavioral and hormonal thoroughly, although a large, placebo-controlled multicen-
responses,13,14 improves ventilator synchrony,15 alleviates ter trial comparing both morphine and fentanyl on impor-
postoperative pain,13,16e18 sedates ventilated preterm tant outcome measures is not available. The neonatal pain
neonates,19,20 and alleviates acute pain caused by invasive consensus statement advises neonatal intensive care units
procedures21e23 (although with variable results24). (NICUs) to use only one opioid analgesic agent to ensure
familiarity with its use.25,26 Clinicians can switch from one
Postnatal intensive care opioid agent to another after prolonged opioid use, or to
obtain better sedation, or to avoid specific side effects,
One of the most frequent applications of opioids in preterm
and term-born neonates is pain treatment during routine
intensive care. According to previous consensus statements
for neonatal pain therapy,25,26 opioids should be considered Table 2 Opioid dosing: starting dosage regimen
for severely painful procedures (Table 1) or as continuous Opioid Bolus/ Continuous
infusions during mechanical ventilation. Continuous mor- intermittent doses infusion dose
phine infusions can be considered for ongoing pain in the Morphine 0.05e0.1 mg/kg iva 0.01e0.03 mg/kg per h
newborn resulting from different etiologies. Fentanyl 0.5e2.0 mg/kg iv 0.5e2.0 mg/kg per h
The clinical effects of the administered opioids should be a
monitored continuously for signs of respiratory or hemody- Evidence for side-effects and worse outcomes were related
with additional morphine boluses given to neonates receiving
namic compromise, or other side effects. The doses listed in
continuous infusions of morphine.
Table 2 are the initial doses recommended for opioid-naive
262 S.H.P. Simons, K.J.S. Anand

such as those related to morphine-mediated histamine re- invasive procedures, such as heel-sticks24 or endotracheal
lease.9,10,27,28 The evidence for opioid cross-tolerance is suctioning.33 Therefore, pre-emptive continuous opioid in-
lacking in neonates, although a limited role of methadone fusions should not be given to newborns routinely during
therapy for treating opioid tolerance and withdrawal has postnatal intensive care. Results from two recently
been proposed.31 performed placebo-controlled randomized trails are
A recent Cochrane meta-analysis of all high-quality summarized in Table 3.
randomized controlled trials studying opioids during neo- Meta-analysis of the data from these two trials, expanded
natal intensive care concluded that insufficient evidence with data from the NOPAIN pilot study, revealed that
was available for routine opioid administration during morphine infusions reduce pain (although the clinical sig-
ventilatory support.32 This was mainly attributed to the nificance of this decrease in pain can be questioned) but
lack of overall benefits from morphine therapy on impor- increase the number of ventilated days.2 Results from the
tant outcome measures such as death, intraventricular NEOPAIN multicenter trial showed that intermittent
hemorrhage, and periventricular leucomalacia. Further- administration of morphine boluses were related to worse
more, continuous morphine administration does not result pulmonary and neurological outcomes and increased hypo-
in clinically significant decreases of pain scores during tension.34e36 Taken together, these data favor the use of

Table 3 Comparison of the NEOPAIN and Dutch trial: studying continuous morphine in newborns receiving ventilatory support
Anand et al.34 Simons et al.33
Methods
Design RCT, placebo controlled, blinded, 16 RCT, placebo controlled, blinded, two centers
centers
Morphine doses Loading dose: 100 mg/kg morphine Loading dose: 100 mg/kg; morphine infusions
infusions 10e30 mg/kg (increasing with GA) 10 mg/kg/h
Additional ‘open label’ morphine permitted Additional ‘open label’ morphine permitted
if necessary if necessary
Primary outcome Composite outcome: neonatal death, Analgesia
severe IVH and PVL
Secondary outcome Analgesia, side-effects Composite outcome, (nor)adrenaline plasma
levels, side-effects
Patients
Inclusion Ventilated, within 72 h of birth Ventilated, within 72 h of birth
Number 898 150
Gestational age range 23e32 weeks 25e42 weeks
Results
Composite outcome Overall: no significant difference. Patients Overall: No significant difference. Significantly
without open label morphine: morphine group more IVH (all stages) in the placebo group
more composite outcome and severe IVH.
Placebo group with ‘open-label’
morphine: more composite outcome.
Morphine group with ‘open label’
morphine: more severe IVH
Analgesia Lower PIPP scores in morphine group No difference in pain scores (PIPP/NIPS/VAS/
after 24 h. No differences in DAN and Comfort score)
PIPP during heelsticks24
‘Open label’ morphine Fewer neonates in morphine group (45.3%) Non-significant difference between morphine
use than in placebo group (54.6%) group (27%) and placebo group (40%)
(Nor)adrenaline plasma e No difference in adrenaline and lower
levels noradrenaline in morphine group37
Side-effects
Hypotension Associated with pre-existing hypotension No overall effect of morphine on blood pressures.
and additional morphine doses35 More frequent hypotension by preemptive
morphine and by ‘open label’ use in
the placebo group38
Pulmonary outcome Morphine group longer ventilated, No differences in length of ventilation
no further differences36
DAN, Douleur Aigue Nouveau-ne; GA, gestational age; IVH, intraventricular haemorrhage; NEOPAIN, neurologic outcomes and pre-
emptive analgesia in neonates; NIPS, Neonatal Infant Pain Scale; PIPP, premature infant pain profile; PVL, periventricular leucomalacia;
VAS, visual analogue scale.
Pain control 263

continuous or slow morphine infusions above the use of re- account.4 An important ethical limitation is the number of
peated, intermittent boluses. Continuous opioid administra- samples and amount of blood that can be obtained for phar-
tion should not be used as the standard of care but started macokinetic analyses of a drug. Also, blood samples cannot
only if required for specific clinical situations in individual always be taken at predetermined times because of clinical
patients. After careful evaluation of the available evidence, constraints. The heterogeneity of the studied population
the NICHD/FDA Pain Control Group recently concluded that adds to this, with widely variable gestational ages, post-
there are limited data to guide clinical analgesic therapy natal ages, bodyweights, variability in severity of illness,
in neonates, and that much more research is needed.1e4 kidney and liver function, amongst other factors.
A modern analysis technique to deal with these compli-
Opioids for surgery or postoperative analgesia cated issues is to analyze data using non-linear mixed-
effects modeling (NONMEM) methods. NONMEM is an
In the 1980s, studies by Anand and Aynsley-Green provided analysis technique that is used in adults to analyze the
evidence for the importance of analgesia during and after kinetics of pharmacological agents. It can be used to provide
neonatal surgery. It was shown that neonates mount a sub- estimates of pharmacokinetic parameters and the explora-
stantial stress response during surgery and that those who tion of covariate relationships (so-called fixed effects).
received fentanyl during surgery had improved postopera- Furthermore, population parameter variability, within and
tive outcome.7 These studies, only 20 years ago, were between subjects, and residual variability (random effects)
a turning-point for many centers around the world to start can be easily incorporated into the model, features that
giving adequate analgesia and anesthesia for neonatal sur- beleaguered the analysis of classical pharmacokinetic
gery. Since then, many studies have been done to improve models. Although originally used for pharmacokinetic anal-
neonatal analgesia during and after surgical procedures. yses, NONMEM can also be used to study the pharmacody-
For postoperative analgesia, again morphine and fen- namics of a drug. Next to plasma levels of opioids and their
tanyl are the best studied and most frequently used opioids, metabolites, the pain scores or stress hormone levels can
although several other agents have been investigated.3,39 also be included into the model. All available PK/PD studies
Many pharmacokinetic and some pharmacodynamic studies of neonatal morphine use were unable to correlate plasma
with morphine have been performed. Clinical trials studying levels with pain scores until now.13,40,44 NONMEM provides
morphine for postoperative analgesia have shown large in- promising opportunities to further analyze the relationships
terindividual variability in morphine plasma levels and between opioid plasma levels and pain scores in newborn
a wide range of morphine requirements.40 Comparing inter- infants. NONMEM has already been used to study
mittent and continuous morphine dosing revealed no,17 or pharmacokinetics in newborns for caffeine, midazolam,
limited,40 differences in pain relief or stress-hormone acetaminophen (paracetamol), antibiotics (such as genta-
levels.14 A meta-analysis calculated that an initial morphine micin and vancomycin), and other drugs used in the neo-
dose of 7 mg/kg/h is sufficient for postoperative analgesia in nate. Only few trials have used NONMEM to analyze
term-born neonates.41 It has been suggested that neonates opioids in newborn infants. Bouwmeester and colleagues
need less morphine after cardiac surgery than after non- analyzed morphine and its metabolites M3G and M6G in new-
cardiac surgery42 and pharmacokinetic studies showed borns and young infants after surgery.48 A mathematical
that morphine requirements increase with gestational and model was created in which a certain dose of morphine
postnatal age.13,43,44 The latter is probably mediated by given results in a plasma morphine concentration with a
immature morphine metabolism, mainly hepatic glucuroni- certain volume of distribution (Fig. 1).
dation from morphine into morphine-3-glucuronide (M3G) Morphine is metabolized in the liver to M6G and M3G and
and the active metabolite morphine-6-glucuronide (M6G), consequently ‘cleared’ into these two metabolites (CL2M6G
both of which are catalyzed by the enzyme UGT2B7. and CL2M3G). Another part of morphine is excreted un-
In general, the doses as listed in Table 2 can also be used metabolized, mainly in the urine (CLEX). The liver-produced
for postoperative opioid analgesia. Perioperative pain con- metabolites result in certain M3G and M6G plasma levels in
trol should, in most cases, begin with the anesthetic man- their specific volumes of distribution (V6M and V3M), and are
agement during surgery. Again postoperatively, opioid excreted mainly in the urine, defined as elimination clear-
doses should be adapted towards the needs and clinical re- ance (CLM6G and CLM3G). The mathematical formulae
sponses of the individual infant. The management of post- underlying this pharmacokinetic model enabled Bouw-
operative pain was also complicated by the lack of meester et al to analyze the effects of age and other factors
objective measurement tools for neonatal pain, a serious on morphine and metabolite pharmacokinetics. A second
limitation in assessing analgesic efficacy until recently.45e47 study plotted Bouwmeester’s data with plasma levels from
neonates receiving various doses of morphine for analgesia
and sedation during extracorporeal membrane oxygenation
Population kinetics (ECMO).49 Interpretation of the morphine plasma levels
from ECMO-treated infants is complicated because of the
Although a number of analgesic trials have been published large ranges in morphine dosing between individuals,
during the last few years, large gaps remain in the current many alterations in dosing for each patient, interactions
pharmacokinetic and pharmacodynamic knowledge of opi- with several co-medications, and unknown effects of
oid analgesics in the newborn infant. Prospective clinical the ECMO membranes. Despite this complexity, using
trials in newborn infants are difficult to perform, time population pharmacokinetics, the clearances of morphine
consuming, expensive and research methods are limited by and its metabolites could be calculated and compared
the special ethical considerations that must be taken into with data from non-ECMO treated newborns (Fig. 2).
264 S.H.P. Simons, K.J.S. Anand

Side effects

The ideal opioid drug would have a rapid onset of action,


a short duration of action, proven efficacy against acute
and chronic pain, and minimal side effects such as hypo-
tension, respiratory depression, chest-wall rigidity, urinary
retention, nausea/vomiting, constipation, increased risk of
necrotizing enterocolitis, and others (Table 4). Long-acting
drugs such as morphine and methadone might not be useful
when immediate analgesia is required, whereas short-
acting drugs like fentanyl, alfentanil and remifentanil can
cause significant hypotension, chest-wall rigidity, and toler-
ance. In addition, very little is known about the side effects
of repeated opioid administration, or specific safety issues
related to critically ill, neurologically impaired, congeni-
tally malformed, or other special populations of neonates.
Figure 1 Pharmacokinetic model of morphine (from ref.,48 One of the main arguments against the treatment of
with permission). CL2M3G/CL2M6G, formation clearance to neonatal pain with opioids is the uncertainty about their
metabolites; CLEX, unaccounted clearance; CLM3G/CLM6G, side effects.50 Research groups studying neuronal apoptosis
elimination clearance of metabolites; CM3G/CM6G, serum me- in rodent models concluded that anesthetic agents are
tabolite concentration; CS, morphine serum concentration; harmful for the neonatal rat brain,51e54 and that these
Dose, dose given; V, volume of distribution for morphine; data might be translated to the human developing
V3M/V6M, volume of distribution of metabolites. brain.53,55 However, repeated administration of high doses
was used in these rodent studies56,57 and their translation
to the human newborn has been questioned.58,59 Neverthe-
This study illustrates the usefulness of NONMEM methods
less, these data continue to support some clinicians in their
for studying the pharmacokinetics of opioids, to calculate
fear of using these anesthetic agents.
clearances and half-lives, and to develop recommendations
Limited data are currently available about the long-term
for opioid doses that should be used under specific circum-
effects of morphine and fentanyl in newborn rats,60e64
stances, such as interactions with comedications or ECMO
whereas the limited evidence available from human trials
systems. The modeling of plasma levels can provide data for
concerning opioid use in the neonatal period generally sug-
mean pharmacokinetic parameters in a specific population
gest beneficial long-term effects.65e67 Large, randomized
of newborn patients, which, in turn, can be used to
controlled trials studying the effects of opioids in human neo-
calculate mean effective analgesic plasma concentrations
natal clinical practice are not able to show negative effects of
and to design a dosing regimen. The interindividual vari-
this treatment.33,34 The expected side effects of morphine,
ability between patients, however, results in the limited
such as hypotension35,38 and effects on pulmonary out-
usefulness of population pharmacokinetics to predict the
come,36 were found to be limited to specific subgroups, and
amount of analgesia required by an individual patient. If
therefore can be avoided. The primary outcomes of the trials
more data become available about DNA polymorphisms
suggest a possible effect of morphine on the development of
explaining the interindividual variability in opioid plasma
intraventricular hemorrhages, which might be a protective
levels between subjects, future studies will incorporate this
effect of routinely administrated morphine,33 or caused by
genetic variability into their PK/PD models.
the intermittent administration of additional morphine
doses.34 Overall intermittent boluses of morphine seem to
be related with worse outcome and side effects.34e36

Table 4 The side-effects of morphine and fentanyl in


newborns
Opioid Side-effect References
Morphine Respiratory depression 41,68e72
Decreased gastrointestinal 9
motility
Hypotension 16,73,74
Urinary retention 75
Risk of necrotizing enterocolitis 76
Figure 2 Individual predicted morphine clearance values, Fentanyl Respiratory depression 6,77
standardized to a 70-kg person, from the NONMEM post hoc Hypotension 78
step, are plotted against age. The solid line demonstrates Muscle rigidity 79e83
a non-linear relationship between clearance and age for post- Hypothermia 7,84
operative children (open diamonds) (from ref., 4 9 with
Adapted from ref.26
permission).
Pain control 265

Side effects from neonatal opioid use on long-term


outcomes remain largely unknown. The follow-up of ran- Research agenda
domized controlled trials that studied effects of morphine
use in the neonatal period did not find any differences in  Despite the information that has become available
the cognitive, behavioral and neuromotor outcomes of from recently performed trials, the safety and
morphine-treated children compared to the control efficacy of short-term and long-term opioid ther-
group.67 This study was flawed by its small sample size apy for newborn patients needs to be evaluated
and the use of pancuronium for some neonates in the in additional well-designed trials.
control group. A non-randomized, retrospective cohort  Pharmacokinetic, dynamic and genetic data
study found that, following birth asphyxia, MRI scans of obtained in from these trials should be analyzed
the neonates who received morphine or fentanyl for post- using non-linear mixed-effects-modeling (NONMEM).
natal intensive care demonstrated significantly less brain  Greater understanding of the genetic, epigenetic,
injury in all regions studied, and these infants had better environmental, and clinical factors that determine
long-term neurologic outcomes.66 The neonates who re- opioid dosing will allow greater predictability of
ceived opioids had also experienced hypoxic/ischemic clinical effects and side effects for individual
insults of greater magnitude (with higher plasma lactate patients.
levels and lower 5-min Apgar scores), suggesting that use
of opioids after perinatal asphyxia might have no significant
long-term detrimental effects and might increase the
brain’s resistance to hypoxic-ischemic damage.66 More
data is required, however, from the follow-up of infants References
that have participated in randomized controlled trials of
opioids during the neonatal period.50,85 1. Anand KJS, Johnston CC, Oberlander T, Taddio A, Tutag-Lehr V,
No large, multicenter, placebo-controlled, randomized Walco GA. Analgesia and local anesthesia during invasive pro-
trial investigating fentanyl in newborns has been per- cedures in the neonate. Clinical Therapeutics 2005;27:
formed. Data from relatively small trials show no serious 844e76.
2. Aranda JV, Carlo WA, Thomas R, Hummel P, Anand KJS. Analge-
side effects on the short time perspective with fentanyl in
sia and sedation during mechanical ventilation in neonates.
newborns.6,30,86 apart from respiratory depression and pro- Clinical Therapeutics 2005;27:877e99.
longed ventilation. Another randomized trial comparing 3. Berde CB, Jaksic T, Lynn AM, Maxwell LG, Soriano SG,
morphine with fentanyl suggests even fewer side effects Tibboel D. Anesthesia and analgesia during and after surgery
from fentanyl than from morphine.9 in neonates. Clinical Therapeutics 2005;27:900e21.
4. Anand KJS, Aranda JV, Berde CB, Buckman S, Capparelli EV,
Carlo WA, et al. Analgesia for neonates: study design and eth-
Conclusions ical issues. Clinical Therapeutics 2005;27:814e43.
5. Yaster M. The dose response of fentanyl in neonatal anesthe-
Clinical trials, mainly including morphine and fentanyl, have sia. Anesthesiology 1987;66:433e5.
shown that opioids can be safely used for neonates during 6. Orsini AJ, Leef KH, Costarino A, Dettorre MD, Stefano JL. Rou-
tine use of fentanyl infusions for pain and stress reduction in
postnatal intensive care and postoperative care. For severe
infants with respiratory distress syndrome. Journal of Pediat-
pain, continuous morphine infusions appear to be the first rics 1996;129:140e5.
choice. During mechanical ventilation and neonatal inten- 7. Anand KJS, Sippell WG, Aynsley-Green A. Randomised trial of
sive care treatment, morphine should not be routinely given fentanyl anaesthesia in preterm babies undergoing surgery:
to all ventilated preterm newborns but can be started for effects on the stress response. Lancet 1987;1:243e8.
the individual infant with significant pain from chest tubes 8. Hickey PR, Hansen DD, Wessel DL, Lang P, Jonas RA, Elixson EM.
or other devices, based on pain scores. Continuous low-dose Blunting of stress responses in the pulmonary circulation of in-
infusions are preferred, particularly for preterm neonates, fants by fentanyl. Anesthesia & Analgesia 1985;64:1137e42.
because intermittent boluses might be related to worse 9. Saarenmaa E, Huttunen P, Leppaluoto J, Meretoja O,
neurological outcomes. The long-term effects of opioids Fellman V. Advantages of fentanyl over morphine in analgesia
for ventilated newborn infants after birth: a randomized trial.
received during the neonatal period remain unknown.
Journal of Pediatrics 1999;134:144e50.
10. Franck LS, Vilardi J, Durand D, Powers R. Opioid withdrawal in
neonates after continuous infusions of morphine or fentanyl
during extracorporeal membrane oxygenation. American Jour-
nal of Critical Care 1998;7:364e9.
Practice points 11. Arnold JH, Truog RD, Scavone JM, Fenton T. Changes in the
pharmacodynamic response to fentanyl in neonates during
 Morphine and fentanyl are reserved for postopera- continuous infusion. Journal of Pediatrics 1991;119:639e43.
12. Arnold JH, Truog RD, Orav EJ, Scavone JM, Hershenson MB. Tol-
tive pain or severe pain during intensive care
erance and dependence in neonates sedated with fentanyl dur-
treatment in newborns. ing extracorporeal membrane oxygenation. Anesthesiology
 During mechanical ventilation, evidence of opioid 1990;73:1136e40.
use is still lacking and should therefore be consid- 13. Bouwmeester NJ, Hop WC, Van Dijk M, Anand KJS, Van Den
ered individually for each patient; doses should be Anker JN, Tibboel D. Postoperative pain in the neonate: age-
adapted to the infant’s needs. related differences in morphine requirements and metabolism.
Intensive Care Medicine 2003;29:2009e15.
266 S.H.P. Simons, K.J.S. Anand

14. Bouwmeester NJ, Anand KJS, van Dijk M, Hop WC, Boomsma F, 31. Chana SK, Anand KJS. Can we use methadone for analgesia in
Tibboel D. Hormonal and metabolic stress responses after neonates? Archives of Disease in Childhood: Fetal & Neonatal
major surgery in children aged 0e3 years: a double-blind, ran- Edition 2001;85:F79e81.
domized trial comparing the effects of continuous versus inter- 32. Bellu R, de Waal KA, Zanini R. Opioids for neonates receiving
mittent morphine. British Journal of Anaesthesia 2001;87: mechanical ventilation. Cochrane Database Systematic
390e9. Reviews 2005;4:CD004212.
15. Dyke MP, Kohan R, Evans S. Morphine increases synchronous 33. Simons SHP, van Dijk M, van Lingen RA, Roofthooft DWA,
ventilation in preterm infants. Journal of Paediatrics & Child Duivenvoorden HJ, Jongeneel N, et al. Routine morphine infu-
Health 1995;31:176e9. sion in preterm newborns who received ventilatory support:
16. Farrington EA, McGuinness GA, Johnson GF, Erenberg A, a randomized controlled trial. JAMA 2003;290:2419e27.
Leff RD. Continuous intravenous morphine infusion in postop- 34. Anand KJS, Hall RW, Desai NS, Shephard B, Bergqvist LL,
erative newborn infants. American Journal of Perinatology Young TE, et al. Effects of pre-emptive morphine analgesia in
1993;10:84e7. ventilated preterm neonates: primary outcomes from the NEO-
17. van Dijk M, Bouwmeester NJ, Duivenvoorden HJ, Koot HM, PAIN trial. Lancet 2004;363:1673e82.
Tibboel D, Passchier J, et al. Efficacy of continuous versus in- 35. Hall RW, Kronsberg SS, Barton BA, Kaiser JR, Anand KJS.
termittent morphine administration after major surgery in 0e Morphine, hypotension and adverse outcomes in preterm neo-
3-year-old infants; a double-blind randomized controlled trial. nates: Who’s to blame? Pediatrics 2005;115:1351e9.
Pain 2002;98:305e13. 36. Bhandari V, Bergqvist LL, Kronsberg SS, Barton BA, Anand KJS.
18. Wolf AR, Hughes D. Pain relief for infants undergoing abdomi- Morphine administration and short-term pulmonary outcomes
nal surgery: comparison of infusions of i.v. morphine and extra- among ventilated preterm infants. Pediatrics 2005;116:352e9.
dural bupivacaine. British Journal of Anaesthesia 1993;70: 37. Simons SH, van Dijk M, van Lingen RA, Roofthooft D,
10e6. Boomsma F, van den Anker JN, et al. Randomised controlled
19. Quinn MW, Wild J, Dean HG, Hartley R, Rushforth JA, trial evaluating effects of morphine on plasma adrenaline/nor-
Puntis JW, et al. Randomized double-blind controlled trial of adrenaline concentrations in newborns. Archives of Disease in
effect of morphine on catecholamine concentrations in venti- Childhood Fetal & Neonatal Edition 2005;90:F36e40.
lated preterm babies. Lancet 1993;342:324e7. 38. Simons SHP, Roofthooft DWE, van Dijk M, van Lingen RA,
20. Wood CM, Rushforth JA, Hartley R, Dean H, Wild J, Levene MI. Duivenvoorden HJ, van den Anker JN, et al. Morphine in venti-
Randomised double blind trial of morphine versus diamorphine lated neonates: its effects on arterial blood pressure. Archives
for sedation of preterm neonates. Archives of Disease in of Disease in Childhood 2006;91:F46e51.
Childhood Fetal & Neonatal Edition 1998;79:F34e9. 39. Berde CB, Sethna NF. Analgesics for the treatment of pain in
21. McCulloch KM, Ji SA, Raju TN. Skin blood flow changes during children. New England Journal of Medicine 2002;347:
routine nursery procedures. Early Human Development 1995; 1094e103.
41:147e56. 40. Lynn AM, Nespeca MK, Bratton SL, Shen DD. Intravenous
22. Moustogiannis AN, Raju TN, Roohey T, McCulloch KM. Intrave- morphine in postoperative infants: intermittent bolus dosing
nous morphine attenuates pain induced changes in skin blood versus targeted continuous infusions. Pain 2000;88:89e95.
flow in newborn infants. Neurological Research 1996;18:440e4. 41. Kart T, Christrup LL, Rasmussen M. Recommended use of mor-
23. Scott CS, Riggs KW, Ling EW, Fitzgerald CE, Hill ML, Grunau RV, phine in neonates, infants and children based on a literature
et al. Morphine pharmacokinetics and pain assessment in pre- review: Part 2eClinical use. Paediatric Anaesthesia 1997;7:
mature newborns. Journal of Pediatrics 1999;135:423e9. 93e101.
24. Carbajal R, Lenclen R, Jugie M, Paupe A, Barton BA, Anand KJS. 42. Lynn A, Nespeca MK, Bratton SL, Strauss SG, Shen DD. Clear-
Morphine does not provide adequate analgesia for acute ance of morphine in postoperative infants during intravenous
procedural pain in preterm neonates. Pediatrics 2005;115: infusion: the influence of age and surgery. Anesthesia &
1494e500. Analgesia 1998;86:958e63.
25. Anonymous. Prevention and management of pain and stress in 43. Kart T, Christrup LL, Rasmussen M. Recommended use of mor-
the neonate. American Academy of Pediatrics. Committee on phine in neonates, infants and children based on a literature
Fetus and Newborn. Committee on Drugs. Section on Anesthe- review: Part 1ePharmacokinetics. Paediatric Anaesthesia
siology. Section on Surgery. Canadian Paediatric Society. Fetus 1997;7:5e11.
and Newborn Committee. Pediatrics 2000;105:454e61. 44. Bouwmeester NJ, van den Anker JN, Hop WC, Anand KJS,
26. Anand KJS. International Evidence-Based Group for Neonatal Tibboel D. Age- and therapy-related effects on morphine re-
Pain. Consensus statement for the prevention and mana- quirements and plasma concentrations of morphine and its
gement of pain in newborns. Archives of Pediatrics and metabolites in postoperative infants. BJA: British Journal of
Adolescent Medicine 2001;155:173e80. Anaesthesia 2003;90:642e52.
27. Ionides SP, Weiss MG, Angelopoulos M, Myers TF, Handa RJ. 45. Barrier G, Attia J, Mayer MN, Amiel-Tison C, Shnider SM.
Plasma beta-endorphin concentrations and analgesia-muscle Measurement of post-operative pain and narcotic administra-
relaxation in the newborn infant supported by mechanical ven- tion in infants using a new clinical scoring system. Intensive
tilation. Journal of Pediatrics 1994;125:113e6. Care Medicine 1989;15:S37e9.
28. Menon G, Anand KJS, McIntosh N. Practical approach to analge- 46. van Dijk M, de Boer JB, Koot HM, Tibboel D, Passchier J,
sia and sedation in the neonatal intensive care unit. Seminars Duivenvoorden HJ. The reliability and validity of the COMFORT
in Perinatology 1998;22:417e24. scale as a postoperative pain instrument in 0 to 3-year old
29. Guinsburg R, Kopelman BI, Anand KJS, de Almeida MF, Peres infants. Pain 2000;84:367e77.
Cde A, Miyoshi MH. Physiological, hormonal, and behavioral 47. McNair C, Ballantyne M, Dionne K, Stephens D, Stevens B. Post-
responses to a single fentanyl dose in intubated and ventil- operative pain assessment in the neonatal intensive care unit.
ated preterm neonates. Journal of Pediatrics 1998;132: Archives of Disease in Childhood Fetal & Neonatal Edition
954e9. 2004;89:F537e41.
30. Lago P, Benini F, Agosto C, Zacchello F. Randomised controlled 48. Bouwmeester NJ, Anderson BJ, Tibboel D, Holford NH. Devel-
trial of low dose fentanyl infusion in preterm infants with hya- opmental pharmacokinetics of morphine and its metabolites
line membrane disease. Archives of Disease in Childhood Fetal in neonates, infants and young children. British Journal of
& Neonatal Edition 1998;79:F194e7. Anaesthesia 2004;92:208e17.
Pain control 267

49. Peters JW, Anderson BJ, Simons SHP, Uges DR, Tibboel D. Mor- 67. MacGregor R, Evans D, Sugden D, Gaussen T, Levene M. Out-
phine pharmacokinetics during venoarterial extracorporeal come at 5e6 years of prematurely born children who received
membrane oxygenation in neonates. Intensive Care Medicine morphine as neonates. Archives of Disease in Childhood Fetal
2005;31:257e63. & Neonatal Edition 1998;79:F40e3.
50. Perlman JM. Morphine, hypotension, and intraventricular hem- 68. Barker DP, Simpson J, Pawula M, Barrett DA, Shaw PN,
orrhage in the ventilated premature infant. Pediatrics 2005; Rutter N. Randomised, double blind trial of two loading dose
115:1416e8. regimens of diamorphine in ventilated newborn infants. Ar-
51. Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vockler J, chives of Disease in Childhood Fetal & Neonatal Edition
Dikranian K, et al. Blockade of NMDA receptors and apoptotic 1995;73:F22e6.
neurodegeneration in the developing brain. Science 1999; 69. Campbell NN, Reynolds GJ, Perkins G. Postoperative analgesia
283:70e4. in neonates: an Australia-wide survey. Anaesthesia & Intensive
52. Jevtovic-Todorovic V, Hartman RE, Izumi Y, Benshoff ND, Care 1989;17:487e91.
Dikranian K, Zorumski CF, et al. Early exposure to common an- 70. Gill AM, Cousins A, Nunn AJ, Choonara IA. Opiate-induced re-
esthetic agents causes widespread neurodegeneration in the spiratory depression in pediatric patients. Annals of Pharmaco-
developing rat brain and persistent learning deficits. Journal therapy 1996;30:125e9.
of Neuroscience 2003;23:876e82. 71. Partridge JC, Wall SN. Analgesia for dying infants whose life
53. Olney JW, Wozniak DF, Jevtovic-Todorovic V, Farber NB, support is withdrawn or withheld. Pediatrics 1997;99:76e9.
Bittigau P, Ikonomidou C. Drug-induced apoptotic neurodegen- 72. Rees EP, Tholl DA. Morphine use and adverse effects in a neona-
eration in the developing brain. Brain Pathology 2002;12: tal intensive care unit. CMAJ (Canadian Medical Association
488e98. Journal) 1994;150:499e504.
54. Olney JW, Farber NB, Wozniak DF, Jevtovic-Todorovic V, 73. Sabatino G, Quartulli L, Di Fabio S, Ramenghi LA. Hemody-
Ikonomidou C. Environmental agents that have the potential namic effects of intravenous morphine infusion in ventilated
to trigger massive apoptotic neurodegeneration in the devel- preterm babies. Early Human Development 1997;47:263e70.
oping brain. Environmental Health Perspectives 2000;108: 74. Goldstein RF, Brazy JE. Narcotic sedation stabilizes arterial
383e8. blood pressure fluctuations in sick premature infants. Journal
55. Olney JW, Young C, Wozniak DF, Ikonomidou C, Jevtovic- of Perinatology 1991;11:365e71.
Todorovic V. Anesthesia induced developmental neuroapopto- 75. Rasch DK, Webster DE, Pollard TG, Gurkowski MA. Lumbar and
sis: does it happen in humans? Anesthesiology 2004;101:530e3. thoracic epidural analgesia via the caudal approach for postop-
56. Hayashi H, Dikkes P, Soriano SG. Repeated administration of erative pain relief in infants and children. Canadian Journal of
ketamine may lead to neuronal degeneration in the developing Anaesthesia 1990;37:359e62.
rat brain. Paediatric Anaesthesia 2002;12:770e4. 76. Hallstrom M, Koivisto AM, Janas M, Tammela O. Frequency of
57. Scallet AC, Schmued LC, Slikker Jr W, Grunberg N, Faustino PJ, and risk factors for necrotizing enterocolitis in infants born be-
Davis H, et al. Developmental neurotoxicity of ketamine: mor- fore 33 weeks of gestation. Acta Paediatrica 2003;92:111e3.
phometric confirmation, exposure parameters, and multiple 77. Vaughn PR, Townsend SF, Thilo EH, McKenzie S, Moreland S,
fluorescent labeling of apoptotic neurons. Toxicological Sci- Denver KK. Comparison of continuous infusion of fentanyl to
ences 2004;81:364e70. bolus dosing in neonates after surgery. Journal of Pediatric
58. Anand KJS, Soriano SG. Anesthetic agents and the immature Surgery 1996;31:1616e23.
brain: are these toxic or therapeutic agents? Anesthesiology 78. Burtin P, Daoud P, Jacqz-Aigrain E, Mussat P, Moriette G. Hypo-
2004;101:527e30. tension with midazolam and fentanyl in the newborn. Lancet
59. Soriano SG, Anand KJS, Rovnaghi CR, Hickey PR. Of mice and 1991;337:1545e6.
men: should we extrapolate rodent experimental data to the 79. Irazuzta J, Pascucci R, Perlman N, Wessel D. Effects of fentanyl
care of human neonates? Anesthesiology 2005;102:866e9. administration on respiratory system compliance in infants.
60. Thornton SR, Smith FL. Characterization of neonatal rat fen- Critical Care Medicine 1993;21:1001e4.
tanyl tolerance and dependence. Journal of Pharmacology & 80. Lemmen RJ, Semmekrot BA. Muscle rigidity causing life-threat-
Experimental Therapeutics 1997;281:514e21. ening hypercapnia following fentanyl administration in a pre-
61. Thornton SR, Wang AF, Smith FL. Characterization of neonatal mature infant. European Journal of Pediatrics 1996;155:1067.
rat morphine tolerance and dependence. European Journal of 81. Lindemann R. Respiratory muscle rigidity in a preterm infant
Pharmacology 1997;340:161e7. after use of fentanyl during Caesarean section. European
62. Thornton SR, Smith FL. Long-term alterations in opiate antino- Journal of Pediatrics 1998;157:1012e3.
ciception resulting from infant fentanyl tolerance and depen- 82. Pokela ML, Ryhanen PT, Koivisto ME, Olkkola KT, Saukkonen AL.
dence. European Journal of Pharmacology 1998;363:113e9. Alfentanil-induced rigidity in newborn infants. Anesthesia &
63. Thornton SR, Lohmann AB, Nicholson RA, Smith FL. Fentanyl Analgesia 1992;75:252e7.
self-administration in juvenile rats that were tolerant and 83. Bolisetty S, Kitchanan S, Whitehall J. Generalized muscle rigid-
dependent to fentanyl as infants. Pharmacology, Biochemistry ity in a neonate following intrathecal fentanyl during caesar-
& Behavior 2000;65:563e70. ean delivery. Intensive Care Medicine 1999;25:1337.
64. Sternberg WF, Scorr L, Smith LD, Ridgway CG, Stout M. Long- 84. Okada Y, Powis M, McEwan A, Pierro A. Fentanyl analgesia in-
term effects of neonatal surgery on adulthood pain behavior. creases the incidence of postoperative hypothermia in neo-
Pain 2005;113:347e53. nates. Pediatric Surgery International 1998;13:508e11.
65. Peters JW, Schouw R, Anand KJS, van Dijk M, Duivenvoorden HJ, 85. Hall RW, Anand KJS. Morphine, hypotension, and intraventric-
Tibboel D. Does neonatal surgery lead to increased pain sensitiv- ular hemorrhage: Response to Dr. Perlman’s commentary. Pe-
ity in later childhood? Pain 2005;114:444e54. diatrics 2005;117, in press.
66. Angeles DM, Wycliffe N, Michelson D, Holshouser BA, 86. Roth B, Schlunder C, Houben F, Gunther M, Theisohn M.
Deming DD, Pearce WJ, et al. Use of opioids in asphyxiated Analgesia and sedation in neonatal intensive care using fen-
term neonates: effects on neuroimaging and clinical outcome. tanyl by continuous infusion. Developmental Pharmacology &
Pediatric Research 2005;57:873e8. Therapeutics 1991;17:121e7.

Você também pode gostar