Você está na página 1de 13

POLICY STATEMENT Organizational Principles to Guide and Define the Child Health

Care System and/or Improve the Health of all Children

Prevention and Management


of Procedural Pain in the
Neonate: An Update
COMMITTEE ON FETUS AND NEWBORN and SECTION ON ANESTHESIOLOGY AND PAIN MEDICINE

The prevention of pain in neonates should be the goal of all pediatricians and abstract
health care professionals who work with neonates, not only because it is
ethical but also because repeated painful exposures have the potential for
deleterious consequences. Neonates at greatest risk of neurodevelopmental
impairment as a result of preterm birth (ie, the smallest and sickest) are
also those most likely to be exposed to the greatest number of painful
stimuli in the NICU. Although there are major gaps in knowledge regarding
the most effective way to prevent and relieve pain in neonates, proven
and safe therapies are currently underused for routine minor, yet painful
procedures. Therefore, every health care facility caring for neonates This document is copyrighted and is property of the American
should implement (1) a pain-prevention program that includes strategies Academy of Pediatrics and its Board of Directors. All authors have
filed conflict of interest statements with the American Academy
for minimizing the number of painful procedures performed and (2) a pain of Pediatrics. Any conflicts have been resolved through a process
approved by the Board of Directors. The American Academy of
assessment and management plan that includes routine assessment of pain, Pediatrics has neither solicited nor accepted any commercial
pharmacologic and nonpharmacologic therapies for the prevention of pain involvement in the development of the content of this publication.

associated with routine minor procedures, and measures for minimizing Policy statements from the American Academy of Pediatrics benefit
from expertise and resources of liaisons and internal (AAP) and
pain associated with surgery and other major procedures. external reviewers. However, policy statements from the American
Academy of Pediatrics may not reflect the views of the liaisons or the
organizations or government agencies that they represent.

The guidance in this statement does not indicate an exclusive course


of treatment or serve as a standard of medical care. Variations, taking
Previous guidance from the American Academy of Pediatrics (AAP) into account individual circumstances, may be appropriate.
and the Canadian Pediatric Society addressed the need to assess
All policy statements from the American Academy of Pediatrics
neonatal pain, especially during and after diagnostic and therapeutic automatically expire 5 years after publication unless reaffirmed,
procedures.1,2 These organizations also provided recommendations revised, or retired at or before that time.
on preventing or minimizing pain in newborn infants and treating DOI: 10.1542/peds.2015-4271
unavoidable pain promptly and adequately.1,2 This statement updates
PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online, 1098-4275).
previous recommendations with new evidence on the prevention,
Copyright © 2016 by the American Academy of Pediatrics
assessment, and treatment of neonatal procedural pain.

To cite: AAP COMMITTEE ON FETUS AND NEWBORN and


BACKGROUND SECTION ON ANESTHESIOLOGY AND PAIN MEDICINE. Prevention
and Management of Procedural Pain in the Neonate: An
Neonates are frequently subjected to painful procedures, with the most
Update. Pediatrics. 2016;137(2):e20154271
immature infants receiving the highest number of painful events.3–5

PEDIATRICS Volume 137, number 2, February 2016:e20154271 FROM THE AMERICAN ACADEMY OF PEDIATRICS
Despite recommendations from the infants; however, multiple factors existing or newly developed tools
AAP and other experts, neonatal interact to influence the nociceptive and against each other to determine
pain continues to be inconsistently processing and/or behavioral which is more reliable for a particular
assessed and inadequately responses to pain.14,16,25–27 Noxious population and application, but more
managed.2,3 A large prospective stimuli activate these signaling research is needed.29,39
study from France in 2008 found pathways but also activate the central Contextual factors such as gestational
that specific pharmacologic or inhibitory circuits, thus altering the age and behavioral state may play a
nonpharmacologic analgesia was balance between the excitatory and significant role in pain assessment
given before painful procedures in inhibitory feedback mechanisms. and are beginning to be included
only 21% of infants, and ongoing The immaturity of the dorsal horn in some assessment tools (eg,
analgesia was given in an additional synaptic connectivity and descending the PIPP-Revised).40,41 New and
34%.3 Thus, infants received inhibitory circuits in neonates emerging technologies to measure
analgesia for approximately half of results in poor localization and pain responses, such as near-infrared
the procedures performed, with wide discrimination of sensory input and spectroscopy, amplitude-integrated
variation among facilities. poor noxious inhibitory modulation, electroencephalography, functional
thus facilitating central nervous MRI, skin conductance, and heart
The prevention and alleviation
system sensitization to repeated rate variability assessment, are being
of pain in neonates, particularly
noxious stimuli.25 investigated.53,54 These innovations
preterm infants, is important not
only because it is ethical but also hold promise in the development
because exposure to repeated painful of neurophysiologically based
ASSESSMENT OF PAIN AND STRESS IN
stimuli early in life is known to THE NEONATE methods for assessing noxious
have short- and long-term adverse stimuli processing at the cortical
Reliable neonatal pain assessment level in neonates while they are
sequelae. These sequelae include
tools are essential for the rating awake, sedated, or anesthetized. If
physiologic instability, altered
and management of neonatal pain, the neurophysiologic measures prove
brain development, and abnormal
and their use has been strongly to be reliable and quantifiable, these
neurodevelopment, somatosensory,
recommended by the AAP and by measures could be used in the future
and stress response systems, which
international researchers, including to simultaneously correlate with
can persist into childhood.5–15
the International Evidence-Based the physiologic and behavioral pain
Nociceptive pathways are active
Group for Neonatal Pain.1,2,28 assessment scales to determine the
and functional as early as 25 weeks’
However, the effective management most clinically useful tool(s).
gestation and may elicit a generalized
of pain in the neonate remains
or exaggerated response to noxious Many of the tools developed to
problematic because of the inability
stimuli in immature newborn measure acute pain in neonates
of the infant to report his or her own
infants.16 are multidimensional in nature and
pain and the challenges of assessing
Researchers have demonstrated that pain in extremely premature, ill, include a combination of physiologic
a procedure-related painful stimulus and neurologically compromised and behavioral signs. These tools
that results in increased excitability neonates.29 Thus, pain assessment were most commonly developed
of nociceptive neurons in the dorsal tools reflect surrogate measures of to assess unventilated infants;
horn of the spinal cord accentuates physiologic and behavioral responses only a few scales are validated
the infant’s sensitivity to subsequent to pain. Although numerous neonatal to assess pain in infants who are
noxious and nonnoxious sensory pain scales exist (Table 1), only 5 ventilated through an endotracheal
stimuli (ie, sensitization).17,18 This pain scales have been subjected to tube or receiving nasal continuous
persistent sensory hypersensitivity rigorous psychometric testing with positive airway pressure.42,55
can be physiologically stressful, the patients serving as their own Recently, investigators reported
particularly in preterm infants.19–22 controls, measuring their physiologic that 2 behaviorally based, one-
Investigators have demonstrated and behavioral responses by using dimensional pain assessment tools
increased stress-related markers the scale in question (Neonatal (the Behavioral Indicators of Infant
and elevated free radicals after even Facial Coding System,30,31 Premature Pain and the Neonatal Facial Coding
simple procedures, such as routine Infant Pain Profile [PIPP],32–34 System) were more sensitive in
heel punctures or tape removal from Neonatal Pain and Sedation Scale,35,36 detecting behavioral cues related
central venous catheters,23,24 which Behavioral Infant Pain Profile,37 and to pain in term neonates than the
can adversely affect future pain Douleur Aiguë du Nouveau-né38). PIPP.56
perception.8 Specific cortical pain Many of the current pain assessment It is unlikely that a single,
processing occurs even in preterm tools have been tested against comprehensive pain assessment

2 FROM THE AMERICAN ACADEMY OF PEDIATRICS


TABLE 1 Pain Assessment Tools for Neonates
Pain Assessment Tool Number and GA of Infants Indicators Intervention Studied Validation Methodology Intended Use
Studied
Neonatal Facial Coding System N = 40 Brow lowering Postoperative abdominal or Patients served as controls Acute pain
(NFCS)30,31 (1998, 2003) 24–32 wk GA Eye squeeze thoracic surgery Interrater reliability: 0.86 Prolonged pain
5–56 DOL Nasolabial furrowing Construct validity: demonstrated Postoperative pain
Lip opening Feasibility: established
Vertical mouth stretch
Horizontal mouth stretch
Taut tongue
Chin quiver
Lip pursing
Premature Infant Pain Profile N = 211, 43, 24 GA Heel lance Patients served as controls Acute pain
(PIPP)32–34 (1996, 1999) Age: 28–40 wk GA Behavioral state Internal consistency: 0.71

PEDIATRICS Volume 137, number 2, February 2016


Maximum HR Construct validity: established
% Decrease in O2 sat Interrater reliability: 0.93–0.96
Brow bulge Intrarater reliability: 0.94–0.98
Eye squeeze
Nasolabial furrow
Neonatal Pain Agitation and N = 42 Crying Heel lance Validated against PIPP Acute pain
Sedation Scale (NPASS)35,36 Age: 23–40 wk GA Behavioral state Interrater reliability: 0.86–0.93 Prolonged pain
(2010) (http://www.n-pass. 1–100 DOL Facial expressions Internal consistency: 0.84–0.89 Level of sedation
com/research.html) Extremities/tone Construct (discriminate) validity:
established
Vital signs (HR, BP, RR, O2 sat) Convergent validity: correlation with the
PIPP scores Spearman rank correlation
coefficient of 0.75 and 0.72
Test-retest reliability: 0.87
Behavioral Indicators of Infant N = 92 Behavioral state Heel lance Validated against NIPS Acute pain
Pain (BIIP)37 (2007) Age: 24–32 wk GA Facial expressions Internal consistency: 0.82
Hand movements Interrater reliability: 0.80–0.92
Construct validity: 85.9
Concurrent validity: correlations between
the BIIP and NIPS = 0.64. Correlations
between the BIIP and mean HR also
remained moderate between GAs:
earlier born = 0.33, P < .05; later born, r
= 0.50, P < .001
Douleur Aiguë du Nouveau-né N = 42 Facial movements Heel lance Patients served as controls Procedural pain
(DAN)38 (1997) Age: 24–41 wk GA Limb movements Venipuncture Internal consistency: 0.88
Vocal expression Interrater reliability: 91.2 (Krippendorf)
Premature Infant Pain Profile– N = 52, 85, 31 Maximum HR Retrospective comparison of Validated against PIPP Acute pain
Revised (PIPP-R)40,41 (2014) Age: 25–40 wk GA % Decrease in O2 sat PIPP and PIPP-R scores Construct validity: established
Brow bulge Feasibility: established
Eye squeeze
Nasolabial furrow
GA and behavioral state assessed if
pain response detected

3
4
TABLE 1 Continued
Pain Assessment Tool Number and GA of Infants Indicators Intervention Studied Validation Methodology Intended Use
Studied
Faceless Acute Neonatal Pain N = 53 HR change Heel lance Validated against DAN Acute pain
Scale (FANS)42 (2010) Age: 30–35 wk GA Acute discomfort (bradycardia, Interrater reliability: 0.92 (0.9–0.98) Developed for use when
desat) the neonate’s face is not
Limb movements Internal consistency: Cronbach’s α = 0.72 completely visible related
Vocal expression (must be The ICC between the FANS and DAN scores to respiratory devices
nonintubated) was 0.88 (0.76–0.93)
Neonatal Infant Pain Scale N = 38 Facial expression Needle insertion Validated against VAS Acute pain
(NIPS)43 (1993) Age: 26–47 wk GA Crying Concurrent validity: correlations with VAS Postoperative pain
ranged from 0.53 to 0.84.
Breathing patterns Interrater reliability: 0.92–0.97
Arm movements Internal consistency: Cronbach’s α’s were
Leg movements 0.95, 0.87, and 0.88 for before, during,
State of arousal and after the procedures, respectively
Crying Requires Increased oxygen N = 24 Crying Postoperative pain Validated against the Objective Pain Score Prolonged pain
administration, Increased Age: 32–60 wk GA Requires O2 to maintain sat at 95% Interrater reliability: 0.72 Postoperative pain
vital signs, Expression, 1382 observations Increased blood pressure, HR Construct validity: yes
Sleeplessness (CRIES)44 (1995) Expression Discriminant validity: yes
Sleep state
COMFORTneo45 (2009) N = 286 Alertness Tertiary NICU care, including Validated against Numeric Rating Scale Persistent or prolonged
ventilation pain
Age: 24.6–42.6 wk GA Calmness/agitation Internal consistency: Cronbach’s α = 0.88 Level of sedation
for nonventilated, 0.84 for ventilated
patients
3600 assessments Respiratory response in ventilated Interrater reliability: 0.79
patient
Crying in spontaneously breathing Concurrent validity: Pearson product-
patient moment correlation coefficient between
COMFORTneo and NRS-pain = 0.54
Body movement Correlation coefficient: 0.75 (95%
Facial tension confidence interval: 0.70–0.79; P <
Body muscle tone .0001)
COVERS Neonatal pain scale46 N = 21 Crying Heel lance Validated different GAs against CRIES, NIPS, Acute pain
(2010) and PIPP
Age: 27–40 wk GA FIO2 requirement Concurrent validity: premature infants
PIPP versus COVERS, r = 0.84; full-term
infants NIPS versus COVERS, r = 0.95
Vital signs (HR, BP, frequency of Construct validity: baseline (P < .05); heel
apnea/bradycardia stick (P < .05); recovery (P < .05)
Facial expression
Resting state
Body movements

FROM THE AMERICAN ACADEMY OF PEDIATRICS


TABLE 1 Continued
Pain Assessment Tool Number and GA of Infants Indicators Intervention Studied Validation Methodology Intended Use
Studied
Pain Assessment in Neonates N = 196 neonates Facial expression Heel lance, suctioning, IV Adapted from NIPS and CRIES Acute pain
(PAIN)47 (2002) Age: 26–47 wk GA Cry placement, circumcision, Inter-rater reliability: not established
Breathing pattern NG tube insertion, tape or IV Correlation between the total scores on
Extremity movement removal the two scales (NIPS and PAIN) was 0.93
State of arousal (P < .001).
FIO2 required for sat >95%
Increase in HR
Pain Assessment Tool (PAT)48,49 N = 144 Posture/tone Ventilated and postoperative Validated against CRIES and VAS Prolonged pain
(2005) Age: 27–40 wk GA Cry neonates Interrater reliability: 0.85
Sleep pattern Correlation between PAT and CRIES scores
Expression (r = 0.76) and (0.38) between the PAT

PEDIATRICS Volume 137, number 2, February 2016


Color score and VAS
Respirations
HR
O2 sat
BP
Nurse’s perception
Scale for Use in Newborns (SUN)50 N = 33 CNS state Intubation Validated against NIPS and COMFORT Acute pain
(1998) Age: 24–40 wk GA Breathing PIV insertion Coefficient of variation: 33 ± 8%
0–214 DOL Movement
68 procedures Tone
Face
HR changes
Mean BP changes
Échelle Douleur Inconfort N = 76 Facial activity Acute and chronic ventilation; Patients served as controls Prolonged pain
Nouveau-Né (EDIN)51 (2001) Age: 25–36 wk GA Body movements NEC, postoperative for PDA Interrater reliability: coefficient range of
ligation 0.59–0.74
Quality of sleep Internal consistency: Cronbach’s α
Quality of contact with nurses coefficients ranged from 0.86 to 0.94
Consolability
Bernese Pain Scale for Neonates N = 12 Alertness Heel lance Validated against VAS and PIPP Acute pain
(BPSN)52 (2004)
Age: 27–41 wk GA Duration of crying Concurrent and convergent validity:
compared with VAS and PIPP was r
= 0.86 and r = 0.91, respectively (P <
.0001)
288 pain assessments Time to calm Interrater reliability: r = 0.86–0.97
Skin color Intrarater reliability: r = 0.98–0.99
Eyebrow bulge with eye squeeze
Posture
Breathing pattern
BP, blood pressure; CNS, central nervous system; desat, desaturation; DOL, days of life; FIO2, fraction of inspired oxygen; GA, gestational age; HR, heart rate; ICC, intraclass correlation coefficient; IV, intravenous (catheter); NEC, necrotizing enterocolitis;
NG, nasogastric; PDA, patent ductus arteriosus; PIV, peripheral intravenous (line); RR, respiratory rate; sat, saturation; VAS, visual analog scale.

5
tool will be satisfactory for assessing preterm neonates and that sucking- or back, and providing oral sucrose
neonatal pain for all situations and related and rocking/holding or glucose solution before a painful
in infants of all gestational ages,39,57 interventions were beneficial for procedure. A systematic review of
although initial validation studies term neonates, but that no benefit 16 studies found that SS was more
have been published for the PIPP- was evident among older infants.64 effective than sucrose when all
Revised in infants with a gestational Skin-to-skin care (SSC), with elements of SS were used,69 and 1
age of 25 to 41 weeks.40,41 More or without sucrose or glucose study suggested that SS may play an
research needs to be performed to administration, has been shown to important role in nonpharmacologic
assess the intensity of both acute decrease some measures of pain management of procedural pain for
and chronic pain at the bedside, to in preterm and term infants.65 An neonates.70
differentiate signs and symptoms analysis of 19 studies examining the
of pain from those attributable to effects of SSC on neonatal pain caused
other causes, and to understand PHARMACOLOGIC TREATMENT
by single needle-related procedures STRATEGIES
the significance of situations when found no statistical benefit for
there is no perceptible response to physiologic indicators of pain but Sucrose and Glucose
pain.40,41 However, even with those did show benefit for composite
limitations, one can use the available Oral sucrose is commonly used
pain score items.65 However, to provide analgesia to infants
evidence to choose a pain assessment some investigators have reported
tool that is appropriate for the type during mild to moderately painful
decreased cortisol concentrations procedures. It has been extensively
of pain assessed (acute, prolonged, and decreased autonomic indicators
postoperative) and advocate for the studied for this purpose, yet
of pain in preterm infants during many gaps in knowledge remain,
competency of the neonatal care SSC, suggestive of a physiologic
provider team with the specific use including appropriate dosing,
benefit.66,67 mechanism of action, soothing versus
of that tool.58 Table 1 lists commonly
used pain assessment tools and the The effects of breastfeeding on analgesic effects, and long-term
evidence used to test them. pain response have also been consequences.71–73 A meta-analysis
investigated. A Cochrane systematic of 57 studies including >4730 infants
review published in 2012 found that with gestational ages ranging from 25
NONPHARMACOLOGIC TREATMENT breastfeeding during a heel lance or to 44 weeks concluded that sucrose
STRATEGIES venipuncture was associated with is safe and effective for reducing
Pediatricians and health care significantly lower pain responses procedural pain from a single event.74
professionals who work with in term neonates (eg, smaller Maximum reductions in physiologic
neonates have the difficult task of increases in heart rate and shorter and behavioral pain indicators
balancing the need for appropriate crying time), compared with other have been noted when sucrose
monitoring, testing, and treatment nonpharmacologic interventions such was administered ∼2 minutes
versus minimizing pain and stress as positioning, rocking, or maternal before a painful stimulus, and the
to the patient. Nonpharmacologic holding. Breastfeeding showed effects lasted ∼4 minutes.74–76
strategies for pain management, similar effectiveness to oral sucrose Procedures of longer duration, such
such as swaddling combined with or glucose solutions.68 This meta- as ophthalmologic examinations or
positioning, facilitated tucking analysis of 20 randomized controlled circumcision, may require multiple
(holding the infant in a flexed trials (RCTs)/quasi-RCTs also found doses of sucrose to provide continual
position with arms close to the trunk) that providing supplemental human analgesic effect.76 In animal studies,
with or without parental assistance, milk via a pacifier or syringe seems to the analgesic effects of sucrose
nonnutritive sucking, and massage, be as effective as providing sucrose appear to be a sweet-taste-mediated
have all shown variable effectiveness or glucose for pain relief in term response of opiate, endorphin, and
in reducing pain and/or stress- neonates. possibly dopamine or acetylcholine
related behaviors related to mild Sensorial stimulation (SS), a method pathways; however, the mechanism
to moderately painful or stressful of gently stimulating the tactile, of action is not well understood in
interventions.59–63 A meta-analysis gustatory, auditory, and visual human neonates.72,77–81 An additive
of 51 studies of nonpharmacologic systems simultaneously, has shown analgesic effect has been noted when
interventions used during heel effectiveness at decreasing pain sucrose is used in conjunction with
lance and intravenous catheter during minor procedures such as other nonpharmacologic measures,
insertion found that sucking-related heel lance.69 SS is achieved by looking such as nonnutritive sucking and
and swaddling/facilitated-tucking at and gently talking to the infant, swaddling, especially for procedures
interventions were beneficial for while stroking or massaging the face such as ophthalmologic examinations

6 FROM THE AMERICAN ACADEMY OF PEDIATRICS


and immunizations.74,78 Although meta-analysis of 38 RCTs that However, effective management
the evidence that oral sucrose included 3785 preterm and strategies for pain and sedation
alleviates procedurally related pain term neonates found that the during mechanical ventilation remain
and stress, as judged by clinical administration of 20% to 30% elusive. A recent systematic review
pain scores, appears to be strong, glucose solutions reduced pain scores reported limited favorable effect with
a small RCT found no difference in and decreased crying during heel selective rather than routine use of
either nociceptive brain activity on lance and venipuncture compared opioids for analgesia in mechanically
electroencephalography or spinal with water or no intervention. The ventilated infants.92 Concerns have
nociceptive reflex withdrawal on authors concluded that glucose been raised for adverse short- and
electromyography between sucrose could be used as an alternative long-term neurodevelopmental
or sterile water administered to term to sucrose solutions, although no outcomes related to the use of
infants before a heel lance.73 This recommendations about dose or morphine infusions in preterm
masked study did find, however, that timing of administration could be neonates.92,93 However, a follow-up
clinical pain scores were decreased made.85 As described for sucrose, study in ninety 8- to 9-year-olds who
in the infants receiving sucrose, and however, glucose may not be had previously participated in 1 RCT
several methodologic concerns limit effective for longer procedures. For comparing continuous morphine
the conclusions that can be drawn example, an RCT found no effect of infusion with placebo found that
from the trial.74 glucose on pain response during low-dose morphine infusion did
ophthalmologic examinations.86 not affect cognition or behavior and
Sucrose use is common in most
may have had a positive effect on
nurseries; however, doses vary
Opioids, Benzodiazepines, and Other everyday executive functions for
widely.82 Although an optimal dose Drugs these children.87
has not been determined,74 an oral
dose of 0.1 to 1 mL of 24% sucrose The most common pharmacologic A 2008 Cochrane systematic
(or 0.2–0.5 mL/kg) 2 minutes agents used for pain relief in review found insufficient evidence
before a painful procedure has been newborns are opioids, with to recommend the routine use of
recommended, taking into account fentanyl and morphine most often opioids in mechanically ventilated
gestational age, severity of illness, used, especially for persistent infants.94 Although there appeared
and procedure to be performed.71 pain. Analgesics and sedatives are to be a reduction in pain, there were
The role and safety of long-term known to be potent modulators no long-term benefits favoring the
sucrose use for persistent, ongoing of several G-protein–linked treatment groups; and concerns for
pain have not been systematically receptor signaling pathways in the adverse effects, such as respiratory
studied. One study in 107 preterm developing brain that are implicated depression, increase in the duration
infants of <31 weeks’ gestation found in the critical regulation of neural of mechanical ventilation, and
worse neurodevelopmental scores tissue proliferation, survival, and development of dependence and
at 32, 36, and 40 weeks’ gestational differentiation. Studies of appropriate tolerance, were raised. Other short-
age in infants who had received >10 dosing and long-term effects of term physiologic adverse effects
doses of sucrose over a 24-hour these analgesics given during the of concern included hypotension,
period in the first week of life, raising neonatal period are woefully lacking constipation, and urinary retention
concerns about frequent dosing in and/or conflicting.87,88 However, for morphine and bradycardia and
newly born preterm infants.83,84 in their absence, it remains critical chest wall rigidity for fentanyl.94
In addition, 1 infant in that study to achieve adequate pain control in Remifentanil, a shorter-acting
developed hyperglycemia coincident newborns, both as an ethical duty fentanyl derivative, may be an
with frequent sucrose dosing, and because painful experiences in alternative for short-term procedures
which may have been related to the the NICU can have long-term adverse and surgeries because it is not
sucrose or to subsequently diagnosed effects.7,10,19,20,89 cleared by liver metabolism, but
sepsis.83 When sucrose is used as a there are no studies examining its
Studies evaluating pharmacologic
pain management strategy, it should long-term effects.95,96
prevention and treatment of mild to
be prescribed and tracked as a
moderate pain have generally been Benzodiazepines, most commonly
medication. More research is needed
limited to a specific procedure such midazolam, are frequently used in the
to better understand the effects of
as intubation. The AAP recommends NICU for sedation. However, because
sucrose use for analgesia.71,81,84
routine pain management during there is evidence of only minor
Glucose has also been found to be procedures such as circumcision,90 additional analgesic effect, they may
effective in decreasing response chest drain insertion and removal,2 not provide much benefit. These
to brief painful procedures. A and nonemergency intubations.91 agents can potentiate the respiratory

PEDIATRICS Volume 137, number 2, February 2016 7


depression and hypotension prolonged hypotension in newborn peripheral arterial puncture.112–114
that can occur with opioids, and infants.105 Limited experience EMLA did not decrease pain-related
infants receiving them should be with dexmedetomidine in preterm measures during heel lance113 but
carefully monitored.97 Midazolam and term infants suggests that it may decrease pain measures during
was associated with adverse may provide effective sedation lumbar puncture,115 particularly if
short-term effects in the NOPAIN and analgesia. Preliminary the patient is concurrently provided
(Neonatal Outcome and Prolonged pharmacokinetic data showed with oral sucrose or glucose
Analgesia in Neonates) trial.98 A decreased clearance in preterm solution.116 Concerns related to the
systematic review in 2012 found infants compared with term infants use of topical anesthetics include
insufficient evidence to recommend and a favorable safety profile over a methemaglobinemia, prolonged
midazolam infusions for sedation 24-hour period.106 application times to allow absorption
in the NICU and raised safety for optimal effectiveness, local skin
The use of oral or intravenous
concerns, particularly regarding irritation, and toxicity, especially in
acetaminophen has been limited
neurotoxicity.97 preterm infants.117,118
to postoperative pain control.
Although intravenous acetaminophen
Alternative medications, such as
has not been approved by the US CONCLUSIONS AND
methadone,99 ketamine, propofol,
Food and Drug Administration, RECOMMENDATIONS
and dexmedetomidine, have been
preliminary data on its safety and
proposed for pain management In summary, there are significant
efficacy are promising in neonates
in neonates; however, few, if any, research gaps regarding the
and infants and it may decrease the
studies of these agents have been assessment, management, and
total amount of morphine needed
performed in this population, and outcomes of neonatal pain; and
to treat postoperative pain.107–109
caution should be exercised when there is a continuing need for studies
Nonsteroidal antiinflammatory
considering them for use because evaluating the effects of neonatal
medication use has been restricted
of concerns about unanticipated pain and pain-prevention strategies
to pharmacologic closure of
adverse effects and potential on long-term neurodevelopmental,
patent ductus arteriosus because
neurotoxic effects.100 Although the behavioral, and cognitive outcomes.
of concerns regarding renal
potential benefits of using methadone The use of pharmacologic treatments
insufficiency, platelet dysfunction,
for the treatment of neonatal pain for pain prevention and management
and the development of pulmonary
include satisfactory analgesic effects in neonates continues to be
hypertension.110 An animal study
and enteral bioavailability as well as hampered by the paucity of data on
suggests that cyclooxygenase-1
prolonged duration of action related the short- and long-term safety and
inhibitors are less effective in
to its long half-life and lower expense efficacy of these agents. At the same
immature compared with mature
compared with other opiates, safe time, repetitive pain in the NICU
animals, probably because of
and effective dosing regimens have has been associated with adverse
decreased cyclooxygenase-1 receptor
yet to be developed.101 Ketamine neurodevelopmental, behavioral,
expression in the spinal cord.110
is a dissociative anesthetic that, and cognitive outcomes, calling for
This decrease in receptor expression
in lower doses, provides good more research to address gaps in
may explain the lack of efficacy of
analgesia, amnesia, and sedation.102 knowledge.5,8,22,89,119–122 Despite
nonsteroidal antiinflammatory drugs
Although ketamine has been well incomplete data, the pediatrician
in human infants.111
studied in older populations, further and other health care professionals
research is needed to establish who care for neonates face the need
Topical Anesthetic Agents
safety profiles for use in neonates to weigh both of these concerns in
because of concerns regarding Topical anesthesia may provide assessing pain and the need for pain
possible neurotoxicity.103 Propofol pain relief during some procedures. prevention and management on a
has been used for short procedural The most commonly studied continuing basis throughout the
sedation in children because of and used topical agents in the infant’s hospitalization.
its rapid onset and clearance. The neonatal population are tetracaine
clearance of propofol in the neonatal gel and Eutectic Mixture of Local Recommendations
population is inversely related to Anesthetics (EMLA), a mixture of 1. Preventing or minimizing pain
postmenstrual age, with significant 2.5% lidocaine and 2.5% prilocaine. in neonates should be the goal
variability in its pharmacokinetics These agents have been found to of pediatricians and other health
in preterm and term neonates.104 decrease measures of pain during care professionals who care
It has also been associated with venipuncture, percutaneous central for neonates. To facilitate this
bradycardia, desaturations, and venous catheter insertion, and goal, each institution should

8 FROM THE AMERICAN ACADEMY OF PEDIATRICS


have written guidelines, based 5. The pediatrician and other health LIAISONS
on existing and emerging care professionals who care for Tonse N.K. Raju, MD, DCH, FAAP – National
evidence, for a stepwise pain- neonates must weigh potential Institutes of Health
prevention and treatment plan, and actual benefits and burdens Captain Wanda D. Barfield, MD, MPH, FAAP –
Centers for Disease Control and Prevention
which includes judicious use of when using pharmacologic
Erin L. Keels, APRN, MS, NNP-BC – National
procedures, routine assessment treatment methods based Association of Neonatal Nurses
of pain, use of both pharmacologic on available evidence. Some Thierry Lacaze, MD – Canadian Pediatric Society
and nonpharmacologic therapies medications can potentiate the Maria Mascola, MD – American College of
for the prevention of pain respiratory depression and Obstetricians and Gynecologists
associated with routine minor hypotension that can occur with
STAFF
procedures, and effective opioids, and infants receiving
Jim Couto, MA
medications to minimize pain them should be carefully
associated with surgery and other monitored. Caution should be SECTION ON ANESTHESIOLOGY AND
major procedures. exercised when considering PAIN MEDICINE EXECUTIVE COMMITTEE,
newer medications for which 2014–2015
2. Despite the significant data in neonates are sparse or Joseph D. Tobias, MD, FAAP, Chairperson
challenges of assessing pain nonexistent. Rita Agarwal, MD, FAAP, Chairperson-Elect
in this population, currently Corrie T.M. Anderson, MD, FAAP
available, validated neonatal 6. Pediatricians, other neonatal Courtney A. Hardy, MD, FAAP
pain assessment tools should be health care providers, and Anita Honkanen, MD, FAAP
consistently used before, during, family members should receive Mohamed A. Rehman, MD, FAAP
continuing education regarding Carolyn F. Bannister, MD, FAAP
and after painful procedures to
monitor the effectiveness of pain the recognition, assessment, and
LIAISONS
relief interventions. In addition, management of pain in neonates,
including new evidence as it Randall P. Flick, MD, MPH, FAAP – American
the need for pain prevention and Society of Anesthesiologists Committee on
management should be assessed becomes available. Pediatrics
on a continuing basis throughout 7. To address the gaps in knowledge, Constance S. Houck, MD, FAAP – AAP Committee
the infant’s hospitalization. on Drugs
more research should be
conducted on pain assessment STAFF
3. Nonpharmacologic strategies,
tools and pharmacologic and
such as facilitated tucking, Jennifer Riefe, MEd
nonpharmacologic strategies
nonnutritive sucking, provision
to prevent or ameliorate pain.
of breastfeeding or providing ABBREVIATIONS
Studies on pharmacokinetics and
expressed human milk, or SS
pharmacodynamics of newer AAP: American Academy of
have been shown to be useful in
medications are needed to prevent Pediatrics
decreasing pain scores during
therapeutic misadventures in PIPP: Premature Infant Pain
short-term mild to moderately
the most vulnerable patients in Profile
painful procedures and should be
pediatric practice. RCT: randomized controlled trial
consistently used.
SS: sensorial stimulation
4. Oral sucrose and/or glucose LEAD AUTHORS SSC: skin-to-skin care
solutions can be effective in Erin Keels, APRN, MS, NNP-BC
neonates undergoing mild to Navil Sethna, MD, FAAP
moderately painful procedures,
either alone or in combination COMMITTEE ON FETUS AND NEWBORN, REFERENCES
with other pain relief strategies. 2015–2016
1. American Academy of Pediatrics
When sucrose or glucose is used Kristi L. Watterberg, MD, FAAP, Chairperson Committee on Fetus and Newborn,
as a pain management strategy, it James J. Cummings, MD, FAAP Committee on Drugs, Section on
should be prescribed and tracked William E. Benitz, MD, FAAP Anesthesiology, Section on Surgery;
as a medication; evidence-based Eric C. Eichenwald, MD, FAAP Canadian Paediatric Society Fetus
protocols should be developed Brenda B. Poindexter, MD, FAAP and Newborn Committee. Prevention
and implemented in nurseries, Dan L. Stewart, MD, FAAP and management of pain and
and more research should be Susan W. Aucott, MD, FAAP stress in the neonate. Pediatrics.
conducted to better understand Jay P. Goldsmith, MD, FAAP 2000;105(2):454–461
the effects of sucrose use for Karen M. Puopolo, MD, PhD, FAAP 2. Batton DG, Barrington KJ, Wallman
analgesia. Kasper S. Wang, MD, FAAP C; American Academy of Pediatrics

PEDIATRICS Volume 137, number 2, February 2016 9


Committee on Fetus and Newborn; primary afferent input and dorsal horn 22. Bouza H. The impact of pain in the
American Academy of Pediatrics cell receptive fields in the rat spinal immature brain. J Matern Fetal
Section on Surgery; Canadian cord is an activity-dependent process. Neonatal Med. 2009;22(9):722–732
Paediatric Society Fetus and Eur J Neurosci. 2002;16(7):1249–1258
23. Bellieni CV, Iantorno L, Perrone S, et
Newborn Committee. Prevention
13. Jennings E, Fitzgerald M. Postnatal al. Even routine painful procedures
and management of pain in the
changes in responses of rat dorsal can be harmful for the newborn. Pain.
neonate: an update. Pediatrics.
horn cells to afferent stimulation: a 2009;147(1–3):128–131
2006;118(5):2231–2241
fibre-induced sensitization. J Physiol. 24. Slater L, Asmerom Y, Boskovic DS,
3. Carbajal R, Rousset A, Danan C, et al. 1998;509(pt 3):859–868
Epidemiology and treatment of painful et al. Procedural pain and oxidative
procedures in neonates in intensive 14. Schmelzle-Lubiecki BM, Campbell stress in premature neonates. J Pain.
care units. JAMA. 2008;300(1):60–70 KA, Howard RH, Franck L, Fitzgerald 2012;13(6):590–597
M. Long-term consequences of 25. Fitzgerald M. The development of
4. Simons SH, van Dijk M, Anand
early infant injury and trauma upon nociceptive circuits. Nat Rev Neurosci.
KS, Roofthooft D, van Lingen RA,
somatosensory processing. Eur J Pain. 2005;6(7):507–520
Tibboel D. Do we still hurt newborn
2007;11(7):799–809
babies? A prospective study of 26. Hohmeister J, Demirakça S, Zohsel
procedural pain and analgesia in 15. Ranger M, Chau CM, Garg A, et al. K, Flor H, Hermann C. Responses to
neonates. Arch Pediatr Adolesc Med. Neonatal pain-related stress predicts pain in school-aged children with
2003;157(11):1058–1064 cortical thickness at age 7 years in experience in a neonatal intensive
5. Anand KJ, Aranda JV, Berde CB, et children born very preterm. PLoS One. care unit: cognitive aspects and
al. Summary proceedings from 2013;8(10):e76702 maternal influences. Eur J Pain.
the neonatal pain-control group. 2009;13(1):94–101
16. Slater R, Cantarella A, Gallella
Pediatrics. 2006;117(3 Suppl 1):S9–S22 27. Grunau RE, Whitfield MF, Fay
S, et al. Cortical pain responses
6. Anand KJ. Clinical importance of pain in human infants. J Neurosci. TB. Psychosocial and academic
and stress in preterm neonates. Biol 2006;26(14):3662–3666 characteristics of extremely low birth
Neonate. 1998;73(1):1–9 weight (< or =800 g) adolescents
17. Ingram RA, Fitzgerald M, Baccei who are free of major impairment
7. Vinall J, Grunau RE. Impact of repeated ML. Developmental changes in the compared with term-born control
procedural pain-related stress in fidelity and short-term plasticity of subjects. Pediatrics. 2004;114(6).
infants born very preterm. Pediatr Res. GABAergic synapses in the neonatal Available at: www.pediatrics.org/cgi/
2014;75(5):584–587 rat dorsal horn. J Neurophysiol. content/full/114/6/e725
8. Doesburg SM, Chau CM, Cheung 2008;99(6):3144–3150
TP, et al. Neonatal pain-related 28. Anand KJ; International Evidence-Based
stress, functional cortical activity 18. Walker SM, Meredith-Middleton Group for Neonatal Pain. Consensus
and visual-perceptual abilities J, Lickiss T, Moss A, Fitzgerald M. statement for the prevention
in school-age children born at Primary and secondary hyperalgesia and management of pain in the
extremely low gestational age. Pain. can be differentiated by postnatal newborn. Arch Pediatr Adolesc Med.
2013;154(10):1946–1952 age and ERK activation in the spinal 2001;155(2):173–180
dorsal horn of the rat pup. Pain.
9. Hermann C, Hohmeister J, Demirakça 29. Hummel P, van Dijk M. Pain
2007;128(1–2):157–168
S, Zohsel K, Flor H. Long-term alteration assessment: current status and
of pain sensitivity in school-aged 19. Holsti L, Grunau RE, Oberlander TF, challenges. Semin Fetal Neonatal Med.
children with early pain experiences. Whitfield MF. Prior pain induces 2006;11(4):237–245
Pain. 2006;125(3):278–285 heightened motor responses 30. Grunau RE, Oberlander T, Holsti L,
during clustered care in preterm Whitfield MF. Bedside application of the
10. Grunau RE, Whitfield MF, Petrie-Thomas
infants in the NICU. Early Hum Dev. Neonatal Facial Coding System in pain
J, et al. Neonatal pain, parenting stress
2005;81(3):293–302 assessment of premature neonates.
and interaction, in relation to cognitive
and motor development at 8 and 20. Grunau RE, Holsti L, Haley DW, et al. Pain. 1998;76(3):277–286
18 months in preterm infants. Pain. Neonatal procedural pain exposure 31. Peters JW, Koot HM, Grunau RE, et
2009;143(1–2):138–146 predicts lower cortisol and behavioral al. Neonatal Facial Coding System
11. Walker SM, Franck LS, Fitzgerald M, reactivity in preterm infants in the for assessing postoperative
Myles J, Stocks J, Marlow N. Long-term NICU. Pain. 2005;113(3):293–300 pain in infants: item reduction
impact of neonatal intensive care and is valid and feasible. Clin J Pain.
21. Cignacco E, Hamers J, van Lingen
surgery on somatosensory perception 2003;19(6):353–363
RA, et al. Neonatal procedural pain
in children born extremely preterm. exposure and pain management in 32. Stevens B, Johnston C, Petryshen P,
Pain. 2009;141(1–2):79–87 ventilated preterm infants during the Taddio A. Premature Infant Pain Profile:
12. Beggs S, Torsney C, Drew LJ, Fitzgerald first 14 days of life. Swiss Med Wkly. development and initial validation. Clin
M. The postnatal reorganization of 2009;139(15–16):226–232 J Pain. 1996;12(1):13–22

10 FROM THE AMERICAN ACADEMY OF PEDIATRICS


33. Ballantyne M, Stevens B, McAllister of validity and reliability. Paediatr in preterm infants. Ann Neurol.
M, Dionne K, Jack A. Validation of Anaesth. 1995;5(1):53–61 2011;70(4):541–549
the premature infant pain profile
45. van Dijk M, Roofthooft DW, Anand 55. Hünseler C, Merkt V, Gerloff M, Eifinger
in the clinical setting. Clin J Pain.
KJ, et al. Taking up the challenge F, Kribs A, Roth B. Assessing pain in
1999;15(4):297–303
of measuring prolonged pain ventilated newborns and infants:
34. Jonsdottir RB, Kristjansdottir G. The in (premature) neonates: the validation of the Hartwig score. Eur J
sensitivity of the Premature Infant COMFORTneo scale seems promising. Pediatr. 2011;170(7):837–843
Pain Profile—PIPP to measure pain Clin J Pain. 2009;25(7):607–616
56. Arias MC, Guinsburg R. Differences
in hospitalized neonates. J Eval Clin
46. Hand I, Noble L, Geiss D, Wozniak L, between uni-and multidimensional
Pract. 2005;11(6):598–605
Hall C. COVERS Neonatal Pain Scale: scales for assessing pain in term
35. Hummel P, Puchalski M, Creech SD, development and validation. Int J newborn infants at the bedside. Clinics
Weiss MG. Clinical reliability and Pediatr. 2010. Available at: www. (Sao Paulo). 2012;67(10):1165–1170
validity of the N-PASS: neonatal hindawi.com/journals/ijpedi/2010/ 57. Ahn Y, Jun Y. Measurement of pain-like
pain, agitation and sedation scale 496719/. Accessed December 17, 2014 response to various NICU stimulants
with prolonged pain. J Perinatol.
47. Hudson-Barr D, Capper-Michel B, for high-risk infants. Early Hum Dev.
2008;28(1):55–60
Lambert S, Palermo TM, Morbeto K, 2007;83(4):255–262
36. Hummel P, Lawlor-Klean P, Weiss MG. Lombardo S. Validation of the Pain 58. Walden M, Gibbins S. Pain Assessment
Validity and reliability of the N-PASS Assessment in Neonates (PAIN) and Management: Guidelines for
assessment tool with acute pain. J scale with the Neonatal Infant Practice. 2nd ed. Glenview, IL: National
Perinatol. 2010;30(7):474–478 Pain Scale (NIPS). Neonatal Netw. Association of Neonatal Nurses; 2010
37. Holsti L, Grunau RE. Initial validation of 2002;21(6):15–21
59. Morrow C, Hidinger A, Wilkinson-Faulk
the Behavioral Indicators of Infant Pain 48. Hodgkinson K, Bear M, Thorn J, D. Reducing neonatal pain during
(BIIP). Pain. 2007;132(3):264–272 Van Blaricum S. Measuring pain in routine heel lance procedures.
38. Carbajal R, Paupe A, Hoenn E, Lenclen neonates: evaluating an instrument MCN Am J Matern Child Nurs.
R, Olivier-Martin M. [APN: evaluation and developing a common language. 2010;35(6):346–354; quiz: 354–356
behavioral scale of acute pain in Aust J Adv Nurs. 1994;12(1):17–22
60. Axelin A, Salanterä S, Kirjavainen J,
newborn infants.] [Article in French]. 49. Spence K, Gillies D, Harrison D, Lehtonen L. Oral glucose and parental
Arch Pediatr. 1997;4(7):623–628 Johnston L, Nagy S. A reliable holding preferable to opioid in pain
39. Cong X, McGrath JM, Cusson RM, Zhang pain assessment tool for clinical management in preterm infants. Clin J
D. Pain assessment and measurement assessment in the neonatal intensive Pain. 2009;25(2):138–145
in neonates: an updated review. Adv care unit. J Obstet Gynecol Neonatal
Nurs. 2005;34(1):80–86 61. Obeidat H, Kahalaf I, Callister LC,
Neonatal Care. 2013;13(6):379–395
Froelicher ES. Use of facilitated
40. Stevens BJ, Gibbins S, Yamada J, 50. Blauer T, Gerstmann D. A simultaneous tucking for nonpharmacological pain
et al. The premature infant pain comparison of three neonatal management in preterm infants: a
profile-revised (PIPP-R): initial pain scales during common systematic review. J Perinat Neonatal
validation and feasibility. Clin J Pain. NICU procedures. Clin J Pain. Nurs. 2009;23(4):372–377
2014;30(3):238–243 1998;14(1):39–47
62. Liaw JJ, Yang L, Katherine Wang KW,
41. Gibbins S, Stevens BJ, Yamada J, et al. 51. Debillon T, Zupan V, Ravault N, Magny Chen CM, Chang YC, Yin T. Non-nutritive
Validation of the Premature Infant Pain JF, Dehan M. Development and initial sucking and facilitated tucking
Profile-Revised (PIPP-R). Early Hum validation of the EDIN scale, a new relieve preterm infant pain during
Dev. 2014;90(4):189–193 tool for assessing prolonged pain in heel-stick procedures: a prospective,
preterm infants. Arch Dis Child Fetal randomised controlled crossover trial.
42. Milesi C, Cambonie G, Jacquot A, Neonatal Ed. 2001;85(1):F36–F41 Int J Nurs Stud. 2012;49(3):300–309
et al. Validation of a neonatal pain
scale adapted to the new practices 52. Cignacco E, Mueller R, Hamers JP, 63. Abdallah B, Badr LK, Hawwari M. The
in caring for preterm newborns. Gessler P. Pain assessment in the efficacy of massage on short and long
Arch Dis Child Fetal Neonatal Ed. neonate using the Bernese Pain term outcomes in preterm infants.
2010;95(4):F263–F266 Scale for Neonates. Early Hum Dev. Infant Behav Dev. 2013;36(4):662–669
2004;78(2):125–131
43. Lawrence J, Alcock D, McGrath P, 64. Pillai Riddell RR, Racine NM, Turcotte
Kay J, MacMurray SB, Dulberg C. 53. Slater R, Cantarella A, Franck L, Meek K, et al. Non-pharmacological
The development of a tool to assess J, Fitzgerald M. How well do clinical management of infant and young child
neonatal pain. Neonatal Netw. pain assessment tools reflect pain in procedural pain. Cochrane Database
1993;12(6):59–66 infants? PLoS Med. 2008;5(6):e129 Syst Rev. 2011;20115(10):CD006275
44. Krechel SW, Bildner J. CRIES: a 54. Smith GC, Gutovich J, Smyser C, et al. 65. Johnston C, Campbell-Yeo M,
new neonatal postoperative pain Neonatal intensive care unit stress is Fernandes A, Inglis D, Streiner D, Zee R.
measurement score. Initial testing associated with brain development Skin-to-skin care for procedural pain

PEDIATRICS Volume 137, number 2, February 2016 11


in neonates. Cochrane Database Syst 77. Fernandez M, Blass EM, Hernandez- and 5-year neurodevelopment outcome
Rev. 2014;1(1):CD008435 Reif M, Field T, Diego M, Sanders in very preterm infants: results from
66. Cong X, Cusson RM, Walsh S, Hussain N, C. Sucrose attenuates a negative the EPIPAGE cohort. Arch Pediatr
Ludington-Hoe SM, Zhang D. Effects of electroencephalographic response to Adolesc Med. 2008;162(8):728–733
skin-to-skin contact on autonomic pain an aversive stimulus for newborns. J 89. Whitfield MF, Grunau RE. Behavior, pain
responses in preterm infants. J Pain. Dev Behav Pediatr. 2003;24(4):261–266 perception, and the extremely low-
2012;13(7):636–645 78. Blass EM, Watt LB. Suckling- and birth weight survivor. Clin Perinatol.
67. Cong X, Ludington-Hoe SM, Walsh sucrose-induced analgesia in human 2000;27(2):363–379
S. Randomized crossover trial of newborns. Pain. 1999;83(3):611–623
90. American Academy of Pediatrics, Task
kangaroo care to reduce biobehavioral 79. Shide DJ, Blass EM. Opioidlike effects Force on Circumcision. Circumcision
pain responses in preterm infants: of intraoral infusions of corn oil policy statement. Pediatrics.
a pilot study. Biol Res Nurs. and polycose on stress reactions 1999;103(3):686–693
2011;13(2):204–216 in 10-day-old rats. Behav Neurosci.
91. Kumar P, Denson SE, Mancuso TJ;
68. Shah PS, Herbozo C, Aliwalas LL, 1989;103(6):1168–1175
Committee on Fetus and Newborn,
Shah VS. Breastfeeding or breast 80. Anseloni VC, Ren K, Dubner R, Ennis M. Section on Anesthesiology and
milk for procedural pain in neonates. A brainstem substrate for analgesia Pain Medicine. Premedication
Cochrane Database Syst Rev. elicited by intraoral sucrose. for nonemergency endotracheal
2012;12(12):CD004950 Neuroscience. 2005;133(1):231–243 intubation in the neonate. Pediatrics.
69. Bellieni CV, Tei M, Coccina F, Buonocore 81. Holsti L, Grunau RE. Considerations for 2010;125(3):608–615
G. Sensorial saturation for infants’ using sucrose to reduce procedural 92. Bellù R, de Waal K, Zanini R. Opioids
pain. J Matern Fetal Neonatal Med. pain in preterm infants. Pediatrics. for neonates receiving mechanical
2012;25(suppl 1):79–81 2010;125(5):1042–1047 ventilation: a systematic review and
70. Gitto E, Pellegrino S, Manfrida M, et 82. Taddio A, Yiu A, Smith RW, Katz J, meta-analysis. Arch Dis Child Fetal
al. Stress response and procedural McNair C, Shah V. Variability in clinical Neonatal Ed. 2010;95(4):F241–F251
pain in the preterm newborn: the practice guidelines for sweetening 93. Anand KJ, Hall RW, Desai N, et al;
role of pharmacological and non- agents in newborn infants undergoing NEOPAIN Trial Investigators Group.
pharmacological treatments. Eur J painful procedures. Clin J Pain. Effects of morphine analgesia
Pediatr. 2012;171(6):927–933 2009;25(2):153–155 in ventilated preterm neonates:
71. Harrison D, Beggs S, Stevens 83. Johnston CC, Filion F, Snider L, et al. primary outcomes from the
B. Sucrose for procedural pain Routine sucrose analgesia during the NEOPAIN randomised trial. Lancet.
management in infants. Pediatrics. first week of life in neonates younger 2004;363(9422):1673–1682
2012;130(5):918–925 than 31 weeks’ postconceptional age. 94. Bellù R, de Waal KA, Zanini R. Opioids
72. Slater R, Cornelissen L, Fabrizi L, et al. Pediatrics. 2002;110(3):523–528 for neonates receiving mechanical
Oral sucrose as an analgesic drug for 84. Johnston CC, Filion F, Snider L, ventilation. Cochrane Database Syst
procedural pain in newborn infants: et al. How much sucrose is too Rev. 2008;1:CD004212
a randomised controlled trial. Lancet. much sucrose [letter]? Pediatrics. 95. Choong K, AlFaleh K, Doucette J, et
2010;376(9748):1225–1232 2007;119(1):226 al. Remifentanil for endotracheal
73. Wilkinson DJ, Savulescu J, Slater 85. Bueno M, Yamada J, Harrison D, et al. intubation in neonates: a randomised
R. Sugaring the pill: ethics and A systematic review and meta-analyses controlled trial. Arch Dis Child Fetal
uncertainties in the use of sucrose for of nonsucrose sweet solutions for pain Neonatal Ed. 2010;95(2):F80–F84
newborn infants. Arch Pediatr Adolesc relief in neonates. Pain Res Manag. 96. Lago P, Tiozzo C, Boccuzzo G, Allegro
Med. 2012;166(7):629–633 2013;18(3):153–161 A, Zacchello F. Remifentanil for
74. Stevens B, Yamada J, Lee GY, Ohlsson 86. Costa MC, Eckert GU, Fortes BG, Fortes percutaneous intravenous central
A. Sucrose for analgesia in newborn Filho JB, Silveira RC, Procianoy RS. catheter placement in preterm infant:
infants undergoing painful procedures. Oral glucose for pain relief during a randomized controlled trial. Paediatr
Cochrane Database Syst Rev. examination for retinopathy of Anaesth. 2008;18(8):736–744
2013;1(1):CD001069 prematurity: a masked randomized 97. Ng E, Taddio A, Ohlsson A. Intravenous
75. Lefrak L, Burch K, Caravantes R, et clinical trial. Clinics (Sao Paulo). midazolam infusion for sedation of
al. Sucrose analgesia: identifying 2013;68(2):199–204 infants in the neonatal intensive care
potentially better practices. Pediatrics. 87. de Graaf J, van Lingen RA, Valkenburg unit. Cochrane Database Syst Rev.
2006;118(2 suppl 2):S197–S202 AJ, et al. Does neonatal morphine 2012;6(6):CD002052
76. Johnston CC, Stremler R, Horton L, use affect neuropsychological 98. Anand KJ, Barton BA, McIntosh N, et
Friedman A. Effect of repeated doses outcomes at 8 to 9 years of age? Pain. al. Analgesia and sedation in preterm
of sucrose during heel stick procedure 2013;154(3):449–458 neonates who require ventilatory
in preterm neonates. Biol Neonate. 88. Rozé JC, Denizot S, Carbajal R, et al. support: results from the NOPAIN trial.
1999;75(3):160–166 Prolonged sedation and/or analgesia Neonatal Outcome and Prolonged

12 FROM THE AMERICAN ACADEMY OF PEDIATRICS


Analgesia in Neonates. Arch Pediatr 107. Allegaert K, van den Anker puncture in newborns. Arch Pediatr
Adolesc Med. 1999;153(4):331–338 J. Pharmacokinetics and Adolesc Med. 2003;157(11):1065–1070
99. Anand KJ. Pharmacological pharmacodynamics of intravenous
116. Biran V, Gourrier E, Cimerman P,
approaches to the management of acetaminophen in neonates. Expert Rev
Walter-Nicolet E, Mitanchez D, Carbajal
pain in the neonatal intensive care Clin Pharmacol. 2011;4(6):713–718
R. Analgesic effects of EMLA cream
unit. J Perinatol. 2007;27(suppl 108. Ceelie I, de Wildt SN, van Dijk M, et al. and oral sucrose during venipuncture
1):S4–S11 Effect of intravenous paracetamol on in preterm infants. Pediatrics.
100. Durrmeyer X, Vutskits L, Anand KJ, postoperative morphine requirements 2011;128(1). Available at: www.
Rimensberger PC. Use of analgesic and in neonates and infants undergoing pediatrics.org/cgi/content/full/128/1/
sedative drugs in the NICU: integrating major noncardiac surgery: a e63
clinical trials and laboratory data. randomized controlled trial. JAMA.
2013;309(2):149–154 117. Foster JP, Taylor C, Bredemeyer SL.
Pediatr Res. 2010;67(2):117–127 Topical anaesthesia for needle-related
101. Chana SK, Anand KJ. Can we use 109. Ohlsson A, Shah PS. Paracetamol pain in newborn infants. Cochrane
methadone for analgesia in neonates? (acetaminophen) for prevention Database Syst Rev. 2013; (1):CD010331
Arch Dis Child Fetal Neonatal Ed. or treatment of pain in newborns.
Cochrane Database Syst Rev. 118. Maulidi H, McNair C, Seller N, Kirsh J,
2001;85(2):F79–F81
2015;6(6):CD011219 Bradley TJ, Greenway SC, Tomlinson
102. Nemergut ME, Yaster M, Colby CE. C. Arrhythmia associated with
Sedation and analgesia to facilitate 110. Ohlsson A, Walia R, Shah S. Ibuprofen
tetracaine in an extremely low birth
mechanical ventilation. Clin Perinatol. for the treatment of patent ductus
weight premature infant. Pediatrics.
2013;40(3):539–558 arteriosus in preterm and/or low birth
2012;130(6). Available at: www.
weight infants. Cochrane Database
103. Cravero JP, Havidich JE. Pediatric pediatrics.org/cgi/content/full/130/6/
Syst Rev. 2008;1:CD003481
sedation—evolution and revolution. e1704
Paediatr Anaesth. 2011;21(7):800–809 111. Ririe DG, Prout HD, Barclay D, Tong
C, Lin M, Eisenach JC. Developmental 119. Harrison D, Yamada J, Stevens B.
104. Allegaert K, Peeters MY, Verbesselt differences in spinal cyclooxygenase Strategies for the prevention and
R, et al. Inter-individual variability in 1 expression after surgical incision. management of neonatal and infant
propofol pharmacokinetics in preterm Anesthesiology. 2006;104(3):426–431 pain. Curr Pain Headache Rep.
and term neonates. Br J Anaesth. 2010;14(2):113–123
2007;99(6):864–870 112. Taddio A, Ohlsson A, Einarson TR,
Stevens B, Koren G. A systematic 120. Grunau R. Early pain in preterm
105. Vanderhaegen J, Naulaers G, Van Huffel review of lidocaine-prilocaine cream infants: a model of long-term effects.
S, Vanhole C, Allegaert K. Cerebral (EMLA) in the treatment of acute pain Clin Perinatol. 2002;29(3):373–394,
and systemic hemodynamic effects in neonates. Pediatrics. 1998;101(2):e1 vii–viii
of intravenous bolus administration
of propofol in neonates. Neonatology. 113. Kapellou O. Blood sampling in infants 121. Taddio A, Shah V, Gilbert-MacLeod C,
2010;98(1):57–63 (reducing pain and morbidity). Katz J. Conditioning and hyperalgesia
2011;Apr 5: 2011. pii: 0313 in newborns exposed to repeated heel
106. Chrysostomou C, Schulman SR,
114. Hall RW, Anand KJ. Pain management lances. JAMA. 2002;288(7):857–861
Herrera Castellanos M, et al A
phase II/III, multicenter, safety, in newborns. Clin Perinatol. 122. Anand KJ, Johnston CC, Oberlander TF,
efficacy, and pharmacokinetic study 2014;41(4):895–924 Taddio A, Lehr VT, Walco GA. Analgesia
of dexmedetomidine in preterm 115. Kaur G, Gupta P, Kumar A. A and local anesthesia during invasive
and term neonates. J Pediatr. randomized trial of eutectic mixture procedures in the neonate. Clin Ther.
2014;164(2):276–282 of local anesthetics during lumbar 2005;27(6):844–876

PEDIATRICS Volume 137, number 2, February 2016 13

Você também pode gostar