Acessibilidade / Reportar erro

Sedation and analgesia for procedures in the pediatric emergency room Please cite this article as: Ramalho CE, Bretas PM, Schvartsman C, Reis AG. Sedation and analgesia for procedures in the pediatric emergency room. J Pediatr (Rio J). 2017;93:2-18. ,☆☆ ☆☆ Study carried out at Universidade de São Paulo (USP), Faculdade de Medicina, Hospital das Clínicas, Instituto da Criança, São Paulo, SP, Brazil.

Abstract

Objective:

Children and adolescents often require sedation and analgesia in emergency situations. With the emergence of new therapeutic options, the obsolescence of others, and recent discoveries regarding already known drugs, it became necessary to review the literature in this area.

Data sources:

Non-systematic review in the PubMed database of studies published up to December 2016, including original articles, review articles, systematic reviews, and meta-analyses. References from textbooks, publications from regulatory agencies, and articles cited in reviews and meta-analyses through active search were also included.

Data synthesis:

Based on current literature, the concepts of sedation and analgesia, the necessary care with the patient before, during, and after sedoanalgesia, and indications related to the appropriate choice of drugs according to the procedure to be performed and their safety profiles are presented.

Conclusions:

The use of sedoanalgesia protocols in procedures in the pediatric emergency room should guide the professional in the choice of medication, the appropriate material, and in the evaluation of discharge criteria, thus assuring quality in care.

KEYWORDS
Conscious sedation; Deep sedation; Analgesia; Child; Emergency service; Emergency medicine

Resumo

Objetivo:

Crianças e adolescentes necessitam frequentemente de sedação e analgesia em situações de emergência. Com o surgimento de novas opções terapêuticas, a obsolescência de outras e descobertas recentes das drogas já conhecidas, fez-se necessário uma nova revisão da literatura nessa área.

Fontes dos dados:

Revisão não sistemática na base de dados PubMed de estudos publicados até dezembro de 2016, inclusive artigos originais, artigos de revisão, revisões sistemáticas e metanálises. Também foram incluídos referências de livros-texto, publicações de agências reguladoras, além de artigos citados nas revisões e metanálises através de busca ativa.

Síntese dos dados:

Com base na literatura atual, são apresentados os conceitos de sedação e analgesia, os cuidados necessários com o paciente antes, durante e após a sedoanalgesia, além de indicações quanto à escolha apropriada dos fármacos de acordo com o procedimento a ser feito e o perfil de segurança desses.

Conclusões:

O emprego de protocolos de sedoanalgesia em procedimentos no pronto-socorro pediátrico deve orientar o profissional na escolha da medicação, do material adequado e na avaliação dos critérios de alta e garantir, assim, qualidade na assistência.

PALAVRAS-CHAVE
Sedação consciente; Sedação profunda; Analgesia; Criança; Serviço de emergência; Medicamento de emergência

Introduction

Children and adolescents often require sedation and analgesia when treated in an emergency situation. Invasive and noninvasive procedures are part of diagnostic and therapeutic techniques in pediatrics, and are often uncomfortable for the child, the parents, and health professionals.11 Krauss B, Green SM. Procedural sedation and analgesia in children. Lancet. 2006;367:766-80. Although necessary, sedation and analgesia may have adverse effects, requiring management in an adequate environment and performed by trained professionals.22 McCoy S, Lyttle MD, Hartshorn S, Larkin P, Brenner M, O'Sullivan R, et al. A qualitative study of the barriers to procedural sedation practices in paediatric emergency medicine in the UK and Ireland. Emerg Med J. 2016;33:527-32.

Sedation for procedures has advanced and expanded in the last decades; no longer the exclusive scope of anesthesiology, now it is being routinely used by the most varied of medical specialties, such as gastroenterology, cardiology, neurology, radiology, emergency medicine, and pediatric intensive care.33 Mason KP. Challenges in paediatric procedural sedation: political, economic, and clinical aspects. Br J Anaesth. 2014;113, Sii48-62. In a qualitative study with physicians from Ireland and the United Kingdom, McCoy et al. identified the lack of training and education in this area as a significant barrier. The standardization of sedation practices and standardization of guidelines and recommendations remains a challenge.44 Gozal D, Mason KP. Pediatric sedation: a global challenge. Int J Pediatr. 2010;:701257. In Brazil, few review articles have been published on this topic in recent years55 Bartolome SM, Cid JL, Freddi N. Analgesia and sedation in children: practical approach for the most frequent situations. J Pediatr (Rio J). 2007;83:S71-82.,66 Miyake RS, Reis AG, Grisi S. Sedation and analgesia for children. Rev Assoc Med Bras. 1998;44:56-64.; protocols are suggested in two of them.66 Miyake RS, Reis AG, Grisi S. Sedation and analgesia for children. Rev Assoc Med Bras. 1998;44:56-64.,77 Carvalho WB, Troster EJ. Sedation and analgesia at the emergency room. J Pediatr (Rio J). 1999;75:S294-306.

Sedation reduces the state of consciousness, while analgesia reduces or eliminates the perception of pain. Many analgesics have some sedative effect, but few sedatives have the property of analgesia. The aim of sedation in pediatrics differs from that in adult patients, as it is administered to control behavior and allow safe completion of the procedure.22 McCoy S, Lyttle MD, Hartshorn S, Larkin P, Brenner M, O'Sullivan R, et al. A qualitative study of the barriers to procedural sedation practices in paediatric emergency medicine in the UK and Ireland. Emerg Med J. 2016;33:527-32. For cooperative children, non-pharmacological modalities should be considered, such as parental presence, hypnosis, distraction, and topical anesthetics, as they may reduce the need for or depth of pharmacological sedation.88 Manyande A, Cyna AM, Yip P, Chooi C, Middleton P. Non-pharmacological interventions for assisting the induction of anaesthesia in children. Cochrane Database Syst Rev. 2015;14:CD006447.,99 Birnie KA, Noel M, Parker JA, Chambers CT, Uman LS, Kisely SR, et al. Systematic review and meta-analysis of distraction and hypnosis for needle-related pain and distress in children and adolescents. J Pediatr Psychol. 2014;39:783-808.

Levels of sedation

Sedation is described as a continuum, represented by progressive stages, from mild to general anesthesia. Ketamine, as an exception, is a dissociative agent that has the particularity of producing a sedation that does not follow this pattern; the effect is present or absent, with maintenance of spontaneous breathing, protective reflexes, and cardiovascular stability.1010 White PF, Way WL, Trevor AJ. Ketamine - its pharmacology and therapeutic uses. Anesthesiology. 1982;56:119-36.

In 2002, the American Society of Anesthesiologists (ASA) defined the four levels of sedation1111. Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology. 2002;96:1004-17.:

  • Minimal sedation (anxiolysis): a state induced by medication in which patients respond normally to verbal commands. Although cognitive function and coordination may be impaired, ventilatory and cardiovascular functions are not affected.

  • Moderate sedation: also called “conscious sedation,” it is a state of drug-induced decrease in consciousness, in which the patient responds purposefully to verbal commands, isolated or accompanied by light tactile stimuli. No intervention is necessary to maintain airway patency and spontaneous ventilation is adequate. In general, cardiovascular function is usually maintained.

  • Deep sedation: a drug-induced decrease in consciousness from which patients cannot be easily awakened, but respond to repeated or painful stimuli. The ability to independently maintain ventilatory function may be impaired. Patients may need assistance to maintain airway patency and spontaneous ventilation may be inadequate. Cardiovascular function is generally maintained.

  • General anesthesia: a state of drug-induced loss of consciousness from which patients are not aroused, even with painful stimuli. The ability to maintain ventilatory function is impaired and patients generally need assistance to maintain airway patency and positive pressure ventilation. Cardiovascular function may be impaired.

One of the important aspects of pain relief in pediatrics implies the understanding of pain assessment methods and their use. Pain can be assessed in children using physiological parameters, behavioral observation, and self-report. The patient with pain will have tachycardia, pupillary dilatation, sweating, and peripheral vasoconstriction.1212 Büttner W, Finke W. Analysis of behavioural and physiological parameters for the assessment of postoperative analgesic demand in newborns, infants and young children: a comprehensive report on seven consecutive studies. Paediatr Anaesth. 2000;10:303-18.,1313 Gonsalves S, Mercer J. Physiological correlates of painful stimulation in preterm infants. Clin J Pain. 1993;9:88-93. No pain assessment should be based solely on these parameters, but rather should be made in combination with validated scales for their adequate measurement. Pain assessment scales have been validated for use in pediatrics, considering the child's developmental phase (verbal or pre-verbal age) and their cognitive ability to report pain.1414 Crellin D, Sullivan TP, Babl FE, O'Sullivan R, Hutchinson A. Analysis of the validation of existing behavioral pain and distress scales for use in the procedural setting. Paediatr Anaesth. 2007;17:720-33.

The Neonatal Infant Pain Scale (NIPS) was developed to evaluate the pain response of patients in the neonatal period, assessing six objective parameters (Table 1).1515 Lawrence J, Alcock D, McGrath P, Kay J, MacMurray SB, Dulberg C. The development of a tool to assess neonatal pain. Neonatal Netw. 1993;12:59-66. The Face, Legs, Activity, Cry, and Consolability (FLACC) scale is validated for children between 2 months and 7 years, and scores five reactions to pain on a scale from 0 to 2 (Table 2).1616 Merkel SI, Voepel-Lewis T, Shayevitz JR, Malviya S. The FLACC: a behavioral scale for scoring postoperative pain in young children. Pediatr Nurs. 1997;23:293-7.,1717 Manworren RC, Hynan LS. Clinical validation of FLACC: preverbal patient pain scale. Pediatr Nurs. 2003;29:140-6. In smaller children, picture-based face are easy to use because they do not require numerical knowledge or certain words (Figs. 1-3).1818 Beyer JE, Aradine CR. Content validity of an instrument to measure young children's perceptions of the intensity of their pain. J Pediatr Nurs. 1986;1:386-95.

19 Bieri D, Reeve RA, Champion GD, Addicoat L, Ziegler JB. The Faces Pain Scale for the self-assessment of the severity of pain experienced by children: development, initial validation, and preliminary investigation for ratio scale properties. Pain. 1990;41:139-50.
-2020 Garra G, Singer AJ, Taira BR, Chohan J, Cardoz H, Chisena E, et al. Validation of the Wong-Baker FACES Pain Rating Scale in pediatric emergency department patients. Acad Emerg Med. 2010;17:50-4. Children older than 8 years are already cognitively able to use visual analog scales (Fig. 4).2121 Todd KH, Funk KG, Funk JP, Bonacci R. Clinical significance of reported changes in pain severity. Ann Emerg Med. 1996;27:485-9.

Table 1
Neonatal Infant Pain Scale (NIPS).
Table 2
Face, Legs, Activity, Cry, and Consolability (FLACC) scale.
Table 3
Classification of the patient's baseline health status, according to the American Society of Anesthesiologists.

Figure 1
OUCHER scale. Adapted from OUCHER.1818 Beyer JE, Aradine CR. Content validity of an instrument to measure young children's perceptions of the intensity of their pain. J Pediatr Nurs. 1986;1:386-95. Explain to the child to score that the intensity of the pain increases in the scale from the bottom up and ask her to point to the figure that demonstrates the intensity of pain she is feeling at the moment.

Figure 2
Faces Pain scale - Revised. Adapted from Faces Pain Scale - Revised © 2001, International Association for the Study of Pain (www.iasp-pain.org/FPSR). Used with permission. Explain to the child to score the chosen face as 0, 2, 4, 6, 8, or 10, counting from left to right; 0 = no pain and 10 = a great deal of pain. Do not use words like “happy” or “sad.” This scale aims to measure how children feel internally and not how they appear to be.

Figure 3
Wong-Baker pain rating scale. Adapted from © 1983 Wong-Baker FACES Foundation. www.WongBakerFACES.org. Use with permission. Explain to the child that each face represents a person with none to a lot of pain. Ask her to choose the face that best translates the pain she is experiencing.

Figure 4
Visual analog scale. Adapted from McCaffery et al.2121 Todd KH, Funk KG, Funk JP, Bonacci R. Clinical significance of reported changes in pain severity. Ann Emerg Med. 1996;27:485-9. Simple numerical scale. Ask the patient to indicate the intensity of the current, the best and the worst level of pain in the last 24 h on a scale of 0 (no pain) to 10 (worst pain imaginable).

Sedation and analgesia for procedures should lead to a state of decreased level of consciousness that allows the patient to maintain airway patency independently and continuously. For this purpose, the correct choice of drugs, doses, and forms of administration are important. Younger and severely ill children often require deeper sedation for painful procedures.

Pre-sedation assessment

The American Society of Anesthesiologists (ASA) recommends the classification of patients into six categories, according to their baseline health (Table 3). Patients classified as ASA I and II are suitable candidates for minimum, moderate, or deep sedation, whereas ASA III and IV patients, those with special needs or anatomic airway abnormalities, require additional considerations, implying moderate or deep sedation.1111. Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology. 2002;96:1004-17. Through the mnemonic acronym “SAMPLE” it is possible to recall the essential components of the patient's medical history that should be considered for sedation assessment (Table 4).

Table 4
Brief and systematic evaluation of the patient submitted to sedation ("SAMPLE").

Adverse events of the airway, cardiovascular system, and respiratory system are the main causes of morbidity and mortality associated with sedoanalgesia in the pediatric population.2222 Tobias JD, Leder M. Procedural sedation: a review of sedative agents, monitoring, and management of complications. Saudi J Anaesth. 2011;5:395-410. A meta-analysis (2016) including 41 studies with 13,883 procedures under sedation in children identified vomiting, restlessness, hypoxia, and apnea as the most frequent complications. In that study, the incidence of severe respiratory adverse events (laryngospasm and need for intubation) was less than 0.5% and respiratory depression occurred in 1.5% of the sedations, with no reports of bronchoaspiration; 97% of laryngospasm cases were associated with the use of ketamine.2323 Bellolio MF, Puls HA, Anderson JL, Gilani WI, Murad MH, Barrionuevo P, et al. Incidence of adverse events in paediatric procedural sedation in the emergency department: a systematic review and meta-analysis. BMJ Open. 2016;6:e011384. Cravero et al. published a multicenter study reporting even lower rates of adverse events, with an incidence of laryngospasm and aspiration of 0.3 and 4.3 per 10,000 cases, respectively.2424 Cravero JP, Blike GT, Beach M, Gallagher SM, Hertzog JH, Havidich JE, et al. Incidence and nature of adverse events during pediatric sedation/anesthesia for procedures outside the operating room: report from the Pediatric Sedation Research Consortium. Pediatrics. 2006;118:1087-96. A meta-analysis of 9652 adults undergoing sedation for procedures also found an incidence of adverse reactions similar to that observed in the pediatric population.2525 Bellolio MF, Gilani WI, Barrionuevo P, Murad MH, Erwin PJ, Anderson JR, et al. Incidence of adverse events in adults undergoing procedural sedation in the emergency department: a systematic review and meta-analysis. Acad Emerg Med. 2016;23:119-34.

Sedative agents have the potential to impair airway protective reflexes, particularly during deep sedation, with the risk of pulmonary aspiration representing one of the reasons to proceed with caution and evaluate the time of fasting before performing a procedure (Table 5). The 2011 ASA recommendations are based on the extrapolation of patients submitted to general anesthesia at the surgical center, not consistent with the reality of sedoanalgesia at the emergency room.2626 American Society of Anesthesiologists Committee. Practice guidelines for preoperative fasting and the use of pharmacologic agents to reduce the risk of pulmonary aspiration: application to healthy patients undergoing elective procedures: an updated report by the American Society of Anesthesiologists Committee on Standards and Practice Parameters. Anesthesiology. 2011;114:495-511. As of 2014, the American College of Emergency Physicians (ACEP) started to recommend that sedation for procedures should not be delayed according to the time of fasting, because there is no evidence that it is related to a reduction in the risk of aspiration or vomiting.2727 Godwin SA, Burton JH, Gerardo CJ, Hatten BW, Mace SE, Silvers SM, et al. Clinical policy: procedural sedation and analgesia in the emergency department. Ann Emerg Med. 2014;63:247-258.e18.

28 Thorpe RJ, Benger J. Pre-procedural fasting in emergency sedation. Emerg Med J. 2010;27:254-61.
-2929 Hoffman GM, Nowakowski R, Troshynski TJ, Berens RJ, Weisman SJ. Risk reduction in pediatric procedural sedation by application of an American Academy of Pediatrics/American Society of Anesthesiologists process model. Pediatrics. 2002;109:236-43. Clark et al. demonstrated that patients with shorter fasting times submitted to deep sedation for elective procedures outside the surgical room experienced similar complication rates when compared to those with longer fasting duration.3030 Clark M, Birisci E, Anderson JE, Anliker CM, Bryant MA, Downs C, et al. The risk of shorter fasting time for pediatric deep sedation. Anesth Essays Res. 2016;10:607-12. Despite ACEP's current recommendations, the authors of this article believe that the risk of sedation and the possibility of aspiration should be weighed against the potential benefits of the procedure, since the number of studies in pediatric emergency units is very small.

Table 5
Adequate fasting time for sedation, according to the American Society of Anesthesiologists.

When performing the physical examination, special attention should be given to cardiac, pulmonary, renal, hepatic, and genetic abnormalities that may alter the child's expected response to analgesic and sedative medications.3131 Butler MG, Hayes BG, Hathaway MM, Begleiter ML. Specific genetic diseases at risk for sedation/anesthesia complications. Anesth Analg. 2000;91:837-55.

32 Malviya S, Voepel-Lewis T, Tait AR. Adverse events and risk factors associated with the sedation of children by nonanesthesiologists. Anesth Analg. 1997;85:1207-13.

33 Jirativanont T, Manomayangkul K, Udomphorn Y, Yokubol B, Saguansab A, Kraiprasit K, et al. Incidence and risk factors for adverse events during anesthesiologist-led sedation or anesthesia for diagnostic imaging in children: a prospective, observational cohort study. Asi Biomed. 2017;9:649-58.
-3434 Caperell K, Pitetti R. Is higher ASA class associated with an increased incidence of adverse events during procedural sedation in a pediatric emergency department?. Pediatr Emerg Care. 2009;25:661-4. Some authors have shown an increased risk of adverse events associated with sedation of patients with comorbidities (ASA > 1).3232 Malviya S, Voepel-Lewis T, Tait AR. Adverse events and risk factors associated with the sedation of children by nonanesthesiologists. Anesth Analg. 1997;85:1207-13.

33 Jirativanont T, Manomayangkul K, Udomphorn Y, Yokubol B, Saguansab A, Kraiprasit K, et al. Incidence and risk factors for adverse events during anesthesiologist-led sedation or anesthesia for diagnostic imaging in children: a prospective, observational cohort study. Asi Biomed. 2017;9:649-58.
-3434 Caperell K, Pitetti R. Is higher ASA class associated with an increased incidence of adverse events during procedural sedation in a pediatric emergency department?. Pediatr Emerg Care. 2009;25:661-4. Airway examination should be thorough, with an active search for characteristics that increase the risk of airway obstruction during the procedure, such as micrognathia, macroglossia, significant tonsil hypertrophy, limited airway opening, extreme obesity, short neck, excessive secretion, or decreased airway protective reflexes.1111. Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology. 2002;96:1004-17. Some genetic and congenital diseases that occur with craniofacial malformations require a more cautious approach due to the difficult airway3535 Nargozian C. The airway in patients with craniofacial abnormalities. Paediatr Anaesth. 2004;14:53-9. (Table 6).

Table 6
Genetic and congenital diseases associated with difficult airway.

In a retrospective study with 11,219 children undergoing general anesthesia procedures with tracheal intubation, laryngoscopy was considered difficult in 1.35% of the cases, with a higher rate in children younger than 1 year of age when compared with those older than 1 year, with an incidence of 4.7% and 0.7%, respectively. A higher risk of difficult laryngoscopy was also identified in patients classified as ASA III and IV, with low body mass index (BMI), submitted to oral cavity, maxillofacial, and cardiac surgery.3636 Heinrich S, Birkholz T, Ihmsen H, Irouschek A, Ackermann A, Schmidt J. Incidence and predictors of difficult laryngoscopy in 11,219 pediatric anesthesia procedures. Paediatr Anaesth. 2012;22:729-36. A similar study analyzed a cohort of 102,305 cases of adult patients submitted to general anesthesia and found a difficult laryngoscopy incidence of 4.9%, three-fold that of the general pediatric population.3737 Heinrich S, Birkholz T, Irouschek A, Ackermann A, Schmidt J. Incidences and predictors of difficult laryngoscopy in adult patients undergoing general anesthesia: a single-center analysis of 102,305 cases. J Anesth. 2013;27:815-21.

Patient vital signs should be recorded prior to the start of sedation, after each medication dose, at regular intervals during the procedure, at the end, during the recovery phase, and at hospital discharge.1111. Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology. 2002;96:1004-17. The American Academy of Pediatrics (AAP) recommends that vital signs should be recorded every ten minutes in patients submitted to moderate sedation and every five minutes for those under deep sedation.3838 Coté CJ, Wilson S. Guidelines for monitoring and management of pediatric patients before, during, and after sedation for diagnostic and therapeutic procedures: update 2016. Pediatrics. 2016;138:pii: e20161212.

Excluding the minimal sedation, where the observation of the level of consciousness is sufficient, patient monitoring must be continuous and include a pulse oximeter, cardiac, respiratory, and blood pressure monitor.3838 Coté CJ, Wilson S. Guidelines for monitoring and management of pediatric patients before, during, and after sedation for diagnostic and therapeutic procedures: update 2016. Pediatrics. 2016;138:pii: e20161212. Capnography can be used in association with the pulse oximeter, being capable of detecting apnea before the latter; it is recommended in moderate sedation and is mandatory in deep sedation.1111. Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology. 2002;96:1004-17.,2727 Godwin SA, Burton JH, Gerardo CJ, Hatten BW, Mace SE, Silvers SM, et al. Clinical policy: procedural sedation and analgesia in the emergency department. Ann Emerg Med. 2014;63:247-258.e18.,3838 Coté CJ, Wilson S. Guidelines for monitoring and management of pediatric patients before, during, and after sedation for diagnostic and therapeutic procedures: update 2016. Pediatrics. 2016;138:pii: e20161212.

A meta-analysis from 2011, based on studies with adult patients submitted to sedation for procedures, identified a 17.6-fold greater probability of detecting respiratory depression when monitoring with capnography.3939 Waugh JB, Epps CA, Khodneva YA. Capnography enhances surveillance of respiratory events during procedural sedation: a meta-analysis. J Clin Anesth. 2011;23:189-96. Langhan et al. randomized 154 children submitted to sedation in the emergency department and found that patients monitored with capnography had earlier interventions in hypoventilation episodes, leading to a lower number of episodes of decreased oxygen saturation in relation to the control group.4040 Langhan ML, Shabanova V, Li FY, Bernstein SL, Shapiro ED. A randomized controlled trial of capnography during sedation in a pediatric emergency setting. Am J Emerg Med. 2015;33:25-30.

Special care should be taken when covering the face and trunk of the child, since the observation of mucosal color and rib cage movement becomes impaired. In other situations of impaired observation, such as magnetic resonance imaging (MRI), continuous non-invasive monitoring equipment (cardiac monitor, oximeter, capnography) should be used. In addition to adequate sources and routes to supply oxygen and suction material, it is mandatory to always have an emergency cart at hand with a defibrillator, resuscitation drugs, antidotes, and equipment for difficult airways.1111. Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology. 2002;96:1004-17.

The presence of a trained professional who knows how to recognize airway impairment and intervene to provide ventilatory support is essential.4141 Krauss BS, Krauss BA, Green SM. Procedural sedation and analgesia in children. N Engl J Med. 2014;370:e23. The AAP recommends that one professional be present for patient monitoring and another with training in airway management and suctioning, bag-mask ventilation, vascular access, and cardiopulmonary resuscitation, both with advanced life support training in pediatrics.3838 Coté CJ, Wilson S. Guidelines for monitoring and management of pediatric patients before, during, and after sedation for diagnostic and therapeutic procedures: update 2016. Pediatrics. 2016;138:pii: e20161212. A study of the Pediatric Sedation Research Consortium analyzed data from 131,751 cases of pediatric patient sedation for procedures in hospitals in the United States, observing that there was no statistical difference in rates of severe adverse events when sedation was performed by different specialists.4242 Couloures KG, Beach M, Cravero JP, Monroe KK, Hertzog JH. Impact of provider specialty on pediatric procedural sedation complication rates. Pediatrics. 2011;127:e1154-60. The training of professionals involved in this type of practice is important to decrease adverse events and promote better patient comfort and safety, and can be carried out using traditional models or through simulations.4343 Sauter TC, Hautz WE, Hostettler S, Brodmann-Maeder M, Martinolli L, Lehmann B, et al. Interprofessional and interdisciplinary simulation-based training leads to safe sedation procedures in the emergency department. Scand J Trauma Resusc Emerg Med. 2016;24:97.

44 Farnsworth ST, Egan TD, Johnson SE, Westenskow D. Teaching sedation and analgesia with simulation. J Clin Monit Comput. 2000;16:273-85.

45 Fehr JJ, Chao J, Kuan C, Zhong J. The important role of simulation in sedation. Curr Opin Anaesthesiol. 2016;29:S14-20.

46 Krauss B, Green SM. Training and credentialing in procedural sedation and analgesia in children: lessons from the United States model. Paediatr Anaesth. 2008;18:30-5.
-4747 Zaveri PP, Davis AB, O'Connell KJ, Willner E, Schinasi DA, Ottolini M. Virtual reality for pediatric sedation: a randomized controlled trial using simulation. Cureus. 2016;8:e486.

One of the periods of greatest risk related to sedation is the recovery phase, so monitoring during this period is mandatory. Patients should be eligible for discharge if they wake up easily, talk and sit unaided, are able to follow age-appropriate commands, are hydrated, and show stable cardiovascular function and patent airways. For very young children or those with some cognitive disorder, with difficulty in interaction, the return to the level of pre-sedation responsiveness must be sought.3838 Coté CJ, Wilson S. Guidelines for monitoring and management of pediatric patients before, during, and after sedation for diagnostic and therapeutic procedures: update 2016. Pediatrics. 2016;138:pii: e20161212. The time of recovery to the baseline state varies with the drug and dose used, but most patients can be discharged after 1-2 h. It is recommended that patients not be submitted to activities that require concentration or motor skills in the first hours after recovery from sedation. Caregivers should be instructed to report any adverse events that occur within 24 h of discharge.4141 Krauss BS, Krauss BA, Green SM. Procedural sedation and analgesia in children. N Engl J Med. 2014;370:e23.

Protocols for sedation and analgesia in the emergency room are essential. The implementation of a specific procedure sedation protocol in a Canadian tertiary hospital reduced the median time between sedation administration and discontinuation of patient monitoring from 49 to 19 min, releasing important resources in a high-demand emergency department4848 Maghraby N, Pearson E, Xue X, Colacone A, Afilalo M. What is the impact of the implementation of an evidence based procedural sedation protocol in the emergency department?. J Clin Trials. 2016;6:283. (Table 7).

Table 7
Steps for sedation and analgesia.

Therapeutic arsenal

The choice of medication used during the sedation and analgesia process should consider the criteria related to the procedure to which the patient will be submitted, as well as the criteria related to the baseline state and comorbidities. Table 8 shows the suggested medications for different types of procedure. The main drugs used in the pediatric emergency unit will be described below. Table 9, at the end of the chapter, summarizes the different characteristics of each drug.

Table 8
Sedoanalgesia for emergency procedures.
Table 9
Pharmacological characteristics of drugs used in sedation and analgesia.

Benzodiazepines

Benzodiazepines are hypnotic sedative agents. Their mechanism of action occurs through their inhibitory effect on the central nervous system (CNS). They bind to postsynaptic gamma-aminobutyric acid (GABA) receptors and increase the permeability to chlorine ions, leading to hyperpolarization and stabilization of the neuronal membrane. They have hepatic metabolism and renal excretion. Their pharmacological effects are sedation, hypnosis, anxiety reduction, amnesia, muscle relaxation, and anticonvulsant effects. This group does not have an analgesic effect and should be associated with other agents, such as opioids, if they are used in painful procedures.4949 Blumer JL. Clinical pharmacology of midazolam in infants and children. Clin Pharmacokinet. 1998;35:37-47. The two main drugs of the group used for sedation in procedures are diazepam and midazolam.

Diazepam has been widely used for sedation, but may cause prolonged sedation because it has a long and variable half-life. Its use has been replaced by midazolam, which was introduced to the market due to its varied routes of administration and shorter duration.11 Krauss B, Green SM. Procedural sedation and analgesia in children. Lancet. 2006;367:766-80. Wright et al., in a prospective, multicenter, randomized study, compared diazepam and midazolam for sedation in emergency department procedures, observing that patients receiving midazolam achieved higher levels of early sedation, higher 90-minute score on the alertness scale, less need for a new dose during the procedure, and less pain during infusion.5050 Wright SW, Chudnofsky CR, Dronen SC, Kothari R, Birrer P, Blanton DM, et al. Comparison of midazolam and diazepam for conscious sedation in the emergency department. Ann Emerg Med. 1993;22:201-5.

Midazolam

Midazolam is the most commonly used intravenous sedative in the emergency department for adults and children. Ilkhanipour et al. carried out a survey in 80 emergency residence programs in the United States and found a 82% rate of institutions using midazolam as the drug of choice for sedation of pediatric patients in the emergency department.5151 Ilkhanipour K, Juels CR, Langdorj MI. Pediatric pain control and conscious sedation: a survey of emergency medicine residencies. Acad Emerg Med. 1994;1:368-72. A study conducted in Brazil by Sukys et al. in a pediatric emergency unit found that midazolam was the sedative of choice in 80% of rapid intubation sequences.5252 Sukys GA, Schvartsman C, Reis AG. Evaluation of rapid sequence intubation in the pediatric emergency department. J Pediatr (Rio J). 2011;87:343-9. The rationale for this fact is the rapid action onset, short duration, anterograde amnesia, and a wide variety of administration routes.

Midazolam is metabolized by cytochrome p450; therefore, the first passage effect should be considered depending on the route of administration; additionally, its metabolization may be compromised in the presence of inflammatory processes, hypoxemia, or use of other drugs metabolized by the same route. It has high protein affinity (97%), thus, in the presence of other medications with the same characteristic, there is an increase in the free fraction of circulating midazolam. Its elimination is 80% by the renal route.4949 Blumer JL. Clinical pharmacology of midazolam in infants and children. Clin Pharmacokinet. 1998;35:37-47. Due to these pharmacological characteristics, it should be administered with caution in patients with hepatopathies and nephropathies. The half-life of midazolam may also be increased in neonates, due to the immaturity of kidney and liver function.5353 Notterman DA. Sedation with intravenous midazolam in the pediatric intensive care unit. Clin Pediatr (Phila). 1997;36:449-54.

It can be administered through the oral (PO), rectal (RR), intranasal (IN), intramuscular (IM), and intravenous (IV) routes. The IV presentation (5 mg/mL) may be used in any route of administration, but it has an irritant effect on the mucosa.5454 Griffith N, Howell S, Mason DG. Intranasal midazolam for premedication of children undergoing day-case anesthesia: comparison of two delivery systems with assessment of intra-observer variability. Br J Anesth. 1998;81:865-9. The oral presentation has a lower concentration (2 mg/mL), being more palatable, which results in a better acceptance by the patients according to specialists.5555 Sahyoun C, Kraus B. Clinical implications of pharmacokinetics and pharmacodynamics of procedural sedation agents in children. Curr Opin Pediatr. 2012;24:225-32. Smith et al., in a randomized study with 77 pediatric patients, observed less discomfort when atomized IN lidocaine at 4% was used as a premedication.5656 Smith D, Cheek H, Denson B, Pruitt CM. Lidocaine pretreatment reduces the discomfort of intranasal midazolam administration: a randomized, double-blind, placebo-controlled trial. Acad Emerg Med. 2017;24:161-7. Chiaretti et al., in a study of 46 children submitted to sedation for non-painful procedures, found no discomfort or pain related to the use of IN midazolam when in combination with lidocaine spray as a premedication.5757 Chiaretti A, Barone G, Rigante D, Ruggiero A, Pierri F, Barbi E, et al. Intranasal lidocaine and midazolam for procedural sedation in children. Arch Dis Child. 2011;96:160-3.

The main side effects are hypotension, respiratory depression, and paradoxical effect. Midazolam reduces peripheral vascular resistance and the action of the sympathetic nervous system, which decreases the cardiac output, with implications mainly in hypovolemic patients or in those with cyanotic heart diseases. Respiratory depression is dose-dependent. In combination with opioids, there is a greater risk of hypotension and respiratory depression; on the other hand, the paradoxical effect is reduced in this association.11 Krauss B, Green SM. Procedural sedation and analgesia in children. Lancet. 2006;367:766-80.,5858 Lucas SS, Nasr VG, Ng AJ, Joe C, Bond M, DiNardo JA. Pediatric Cardiac Intensive Care Society 2014 consensus statement: pharmacotherapies in cardiac critical care: sedation, analgesia and muscle relaxant. Pediatr Crit Care Med. 2016;17:S3-15.

Flumazenil

Flumazenil is a central-action benzodiazepine antagonist and its administration is exclusively by the intravenous route. Reversal of benzodiazepine effects is contrary to its appearance during sedation, i.e., small doses of midazolam are necessary to generate an anxiolytic effect, while higher doses of flumazenil are necessary to reverse this effect. In turn, high doses of midazolam are required to induce deep sedation, while small doses of flumazenil reverse this effect. Side effects are elevated intracranial pressure and decreased convulsive threshold, so it should be used with caution in patients taking medications that may induce seizures (tricyclic antidepressants, cocaine, lithium, methylxanthines, isoniazid, monoamine oxidase inhibitors, bupropion, theophylline).5959 Shannon M, Albers G, Burkart K, Liebelt E, Kelley M, McCubbin MM, et al. Safety and efficacy of flumazenil in the reversal of benzodiazepine-induced conscious sedation. J Pediatr. 1997;131:582-6.

Opioids

Opioids modulate the cortical perception of pain. They act by binding to central and peripheral µ, δ, and κ receptors, causing cellular hyperpolarization, reducing the release of neurotransmitters. Their main indication is for relief of moderate to severe pain.1212 Büttner W, Finke W. Analysis of behavioural and physiological parameters for the assessment of postoperative analgesic demand in newborns, infants and young children: a comprehensive report on seven consecutive studies. Paediatr Anaesth. 2000;10:303-18.,1313 Gonsalves S, Mercer J. Physiological correlates of painful stimulation in preterm infants. Clin J Pain. 1993;9:88-93. Morphine and fentanyl are the most commonly used opioids in clinical practice.

Morphine

Morphine is an opioid with a delayed action onset (5-10 min) and prolonged duration (120-180 min). Delay in the clinical effect occurs because of its relatively low lipid solubility. It undergoes significant first passage metabolism and, therefore, oral doses should be six-fold greater than parenteral doses to achieve the same degree of analgesia.6060 Inturrisi CE. Clinical pharmacology of opioids for pain. Clin J Pain. 2002;18:S3-13.,6161 Stanski DR, Greenblatt DJ, Lowenstein E. Kinetics of intravenous and intramuscular morphine. Clin Pharmacol Ther. 1978;24:52-9. It is indicated in procedures in which the aim is to maintain analgesia for a longer time, e.g., in fracture fixation. Barcelos et al. compared morphine (0.1 mg/kg) and ketamine (2 mg/kg), both associated with midazolam (0.2 mg/kg), for analgesia in fracture reduction in 25 children in an emergency department, and found no statistical difference regarding failure rate, procedure start time, length of hospital stay, pain scales, or rate of satisfaction of parents and orthopedists.6262 Barcelos A, Garcia PC, Portela JL, Piva JP, Garcia JP, Santana JC. Comparison of two analgesia protocols for the treatment of pediatric orthopedic emergencies. Rev Assoc Med Bras. 2015;61:362-7. Another recurrent use in clinical practice is the relief of acute or chronic intense pain, such as in patients with sickle-cell disease.6363 Gupta M, Msambichaka L, Ballas SK, Gupta K. Morphine for the treatment of pain in sickle cell disease. Sci World J. 2015;2015:540154.

The metabolism of morphine is hepatic and extrahepatic, with the metabolites being excreted in the urine. In patients younger than 6 months, the metabolization mechanisms are immature, and there is evidence of decreased clearance in children with cardiovascular instability.6161 Stanski DR, Greenblatt DJ, Lowenstein E. Kinetics of intravenous and intramuscular morphine. Clin Pharmacol Ther. 1978;24:52-9.,6464 Martin LD, Jimenez N, Lynn AM. A review of perioperative anesthesia and analgesia for infants: updates and trends to watch. F1000Res. 2017;6:120. McRorie et al. observed that children receiving IV morphine after cardiac surgery reached adult morphine clearance values at 6 months of age.6565 McRorie TI, Lynn AM, Nespeca MK, Opheim KE, Slattery JT. The maturation of morphine clearance and metabolism. Am J Dis Child. 1992;146:972-6. In another study, Lynn et al. identified adult morphine clearance values in infants around 1-3 months of age.6666 Lynn A, Nespeca MK, Bratton SL, Strauss SG, Shen DD. Clearance of morphine in postoperative infants during intravenous infusion: the influence of age and surgery. Anesth Analg. 1998;86:958-63. The variability of morphine duration is high, which makes it difficult to predict the duration of morphine effects.

Morphine stimulates the release of significant amounts of histamine and inhibits compensatory sympathetic responses. This effect can cause bronchoconstriction and is deleterious in asthmatics. Vasodilation produced by histamine may result in hypotension, especially with rapid infusion administration.6767 Rang HP, Dale MM, Ritter JM, Flower RJ, Henderson G. Farmacologia. 7th ed. Rio de Janeiro: Elsevier; 2012.

Important gastrointestinal side effects are also observed with the use of morphine, such as nausea and vomiting that can occur in up to 40% of patients, an effect that tends to decrease with repeated doses of the medication. Another effect, which is common to all opioids, is increased tonus and reduced gastrointestinal motility.6767 Rang HP, Dale MM, Ritter JM, Flower RJ, Henderson G. Farmacologia. 7th ed. Rio de Janeiro: Elsevier; 2012.

Discontinuation of morphine infusion is associated with abstinence phenomena. Signs and symptoms include pupillary dilation, tearing, sweating, shivers, hypertension, fever, vomiting, abdominal pain, diarrhea, muscle and joint pain, and behavioral changes.6868 Tobias JD. Tolerance, withdrawal, and physical dependency after long-term sedation and analgesia of children in the pediatric intensive care unit. Crit Care Med. 2000;28:2122-32.

Fentanyl

Fentanyl is a synthetic opioid with approximately 100 times the analgesic power of morphine. It is highly liposoluble, which explains its rapid action onset. It has a half-life of 2-4 h on intermittent administration and 21 h after prolonged continuous infusion. This difference occurs due to the saturation of opioid receptors in the lipophilic peripheral tissues.6969 Feierman DE, Lasker JM. Metabolism of fentanyl, a synthetic opioid analgesic, by human liver microsomes: role of CYP3A4. Drug Metab Dispos. 1996;24:932-9.,7070 Labroo RB, Paine MF, Thummel KE, Kharasch ED. Fentanyl metabolism by human hepatic and intestinal cytochrome P4503A4: implications for interindividual variability in disposition, efficacy, and drug interactions. Drug Metab Dispos. 1997;25:1072-80.

The metabolism of fentanyl occurs almost exclusively in the liver; it has no active metabolites and a small fraction is excreted unaltered in the urine. Infants and young children have greater clearance than older children and adults, often requiring more frequent doses. Singleton et al. evaluated the plasma concentration of fentanyl in three different age groups and found a higher plasma concentration of fentanyl in adults compared to children, and an even lower concentration in infants.7171 Singleton MA, Rosen JI, Fisher DM. Plasma concentrations of fentanyl in infants, children and adults. Can J Anaesth. 1987;34:152-5.

Some properties of fentanyl have clinical implications, for instance, changes in blood pH can alter its ionization and distribution between plasma and the CNS, and patients with acidosis may have an increase in the free fraction of fentanyl, putting them at higher risk of toxicity.7272 Lexi-Comp Inc. Lexicomp Online, Pediatric and Neonatal Lexi-Drugs Online. Hudson, OH: Lexi-Comp, Inc, April-May 2017; 2017.

Studies have shown that fentanyl administered by alternative routes is effective in relieving pain. Miner et al., in a randomized clinical trial of 41 children who received 1.5 mcg/kg of IV fentanyl or 3 mcg/kg of nebulized fentanyl, showed a similar result in the pain improvement score.7373 Miner JR, Kletti C, Herold M, Hubbard D, Biros MH. Randomized clinical trial of nebulized fentanyl citrate versus i.v. fentanyl citrate in children presenting to the emergency department with acute pain. Acad Emerg Med. 2007;14:895-8. Borland et al., in a study comparing IN fentanyl at a dose of 1.7 mg/kg with IV morphine at a dose of 0.1 mg/kg, showed similar improvement in pain scores in the two groups of pediatric patients submitted to fracture reduction.7474 Borland M, Milsom S, Esson A. Equivalency of two concentrations of fentanyl administered by the intranasal route for acute analgesia in children in a paediatric emergency department: a randomized controlled trial. Emerg Med Aust. 2011;23:202-8. Similar results were achieved by Young et al. when comparing intranasal fentanyl (1 mcg/kg) with IM morphine (0.2 mg/kg) in fracture reduction in pediatric patients, and tolerance to administration was better in the IN fentanyl group.7575 Young PA, Nicol MF, Kendall JM, Harrington AP. A prospective randomized pilot comparison of intranasal fentanyl and intramuscular morphine for analgesia in children presenting to the emergency department with clinical fractures. Emerg Med. 1999;11:90-4.

Fentanyl can cause hemodynamic impairment by bradycardia-induced decreased cardiac output.5858 Lucas SS, Nasr VG, Ng AJ, Joe C, Bond M, DiNardo JA. Pediatric Cardiac Intensive Care Society 2014 consensus statement: pharmacotherapies in cardiac critical care: sedation, analgesia and muscle relaxant. Pediatr Crit Care Med. 2016;17:S3-15. A widely reported adverse effect, although rare, is chest wall stiffness, which is associated with high doses, above 5 mcg/kg with bolus administration.6060 Inturrisi CE. Clinical pharmacology of opioids for pain. Clin J Pain. 2002;18:S3-13.

Respiratory depression is a side effect common to all opioids, and is associated with the administered dose. The incidence of respiratory depression is significantly higher when opioids are used in combination with benzodiazepines. Roback et al. analyzed the occurrence of side effects in 2500 patients who underwent sedation and analgesia in a pediatric emergency department in the United States, and found a rate of respiratory adverse events of 19.3% when there was an association between midazolam and fentanyl, and of 5.8% when midazolam was administered alone.5959 Shannon M, Albers G, Burkart K, Liebelt E, Kelley M, McCubbin MM, et al. Safety and efficacy of flumazenil in the reversal of benzodiazepine-induced conscious sedation. J Pediatr. 1997;131:582-6.,7676 Roback MG, Wathen JE, Bajaj L, Bothner JP. Adverse events associated with procedural sedation and analgesia in a pediatric emergency department: a comparison of common parenteral drugs. Acad Emerg Med. 2005;12:508-13.

The potential advantages of fentanyl over morphine for performing the procedure are: faster onset of action, shorter half-life, and avoidance of histamine release, with a low incidence of nausea, vomiting, and generalized pruritus. Additionally, there is no cross-reaction with morphine allergy.7777 Fleischman RJ, Frazer DG, Daya MJ, Jui J, Newgard CD. Effectiveness and safety of fentanyl compared with morphine for out-of-hospital analgesia. Prehosp Emerg Care. 2010;14:167-75.

Naloxone

Naloxone is an opioid receptor antagonist. The most commonly used administration routes are IV and IM, but it can also be administered by the subcutaneous, sublingual, and endotracheal routes. The dose varies according to the desired effect, i.e., partial reduction of the opioid effect (0.01-0.03 mg/kg) or complete reversal (0.1-2 mg/kg). After its administration, respiration return is observed in one to two minutes, and transient tachypnea may occur. Doses may be repeated every two minutes if the expected effect is not achieved or the reversal has been transient, since the half-life of naloxone is lower than that of opioids.7878 Barsan WG, Seger D, Danzl DF, Ling LJ, Bartlett R, Buncher R, et al. Duration of antagonistic effects of nalmefene and naloxone in opiate-induced sedation for emergency department procedures. Am J Emerg. 1989;7:155-61.

Possible side effects are nausea, anxiety, sympathetic stimulation, hypertension, tachycardia, pulmonary edema, and return of pain. At a low dose, naloxone seems to reduce the nausea caused by opioids. A meta-analysis carried out by Barrons and Woods concluded that the use of naloxone reduces the occurrence of postoperative nausea without significantly increasing the need for extra doses of opioids and without increasing pain scores; however, there was no decrease in the occurrence of vomiting.7979 Barrons RW, Woods JA. Low-dose naloxone for prophylaxis of postoperative nausea and vomiting: a systematic review and meta-analysis. Pharmacotherapy. 2017;37:546-54.

Ketamine

Ketamine is a dissociative anesthetic agent that acts on the N-methyl-D-aspartate-glutamate receptor, disconnecting the limbic and thalamocortical systems, dissociating the central nervous system from external stimuli. The cataleptic state allows potent analgesia, sedation, and amnesia, while maintaining airway patency, protective stimuli, and cardiovascular stability.1010 White PF, Way WL, Trevor AJ. Ketamine - its pharmacology and therapeutic uses. Anesthesiology. 1982;56:119-36.

It is widely used in painful short-term procedures or in those in which amnesia is desired, such as for the physical examination of patients that are victims of abuse.8080 Green SM, Roback MG, Kennedy RM, Krauss B. Clinical practice guideline for emergency department ketamine dissociative sedation: 2011 update. Ann Emerg Med. 2011;57:449-61. It is not recommended for sedation in computed tomography (CT) of the skull or MRI, because the dissociative state can produce inappropriate movements, resulting in poorer image quality. It has hepatic metabolism and predominantly urinary excretion (91%).1010 White PF, Way WL, Trevor AJ. Ketamine - its pharmacology and therapeutic uses. Anesthesiology. 1982;56:119-36.

It is typically administered intravenously or intramuscularly, and may be administered by the intranasal and oral routes. The IV route allows faster recovery and less time until discharge, while the IM route is an independent predictor of emesis.8181 Deasy C, Babl FE. Intravenous vs intramuscular ketamine for pediatric procedural sedation by emergency medicine specialists: a review. Paediatr Anaesth. 2010;20:787-96.,8282 Green SM, Roback MG, Krauss B, Brown L, McGlone RG, Agrawal D, et al. Emergency Department Ketamine Meta-Analysis Study Group. Predictors of emesis and recovery agitation with emergency department ketamine sedation: an individual-patient data meta-analysis of 8,282 children. Ann Emerg Med. 2009;54:171-80. In a randomized study, the administration of higher doses of ketamine (1.5 and 2 mg/kg) compared to a lower dose (1 mg/kg) decreased the need for new doses to achieve adequate sedation (2.9% and 5% vs. 16%), with no increase in the risk of adverse effects (14.3% and 10% vs. 10%) or the duration of sedation in minutes (24.5 min and 23 min vs. 23 min).8383 Kannikeswaran N, Lieh-Lai M, Malian M, Wang B, Farooqi A, Roback MG. Optimal dosing of intravenous ketamine for procedural sedation in children in the ED - a randomized controlled trial. Am J Emerg Med. 2016;34:1347-53.

Ketamine can cause transient laryngospasm, as well as apnea, hypersalivation, vomiting, and restlessness in recovery. It inhibits the reuptake of catecholamines, resulting in a sympathomimetic effect, which causes an increase in blood pressure, heart rate, and cardiac output. A prospective study of 11 adult patients with ischemic heart disease showed a reduction in left ventricular systolic and diastolic function.8484 Jakobsen CJ, Torp P, Vester AE. Ketamine reduce left ventricular systolic and diastolic function in patients with ischaemic heart disease. Acta Anaesthesiol Scand. 2010;54:1137-44. Reduction of systolic pressure secondary to decreased ejection fraction after ketamine use was demonstrated in a small group of patients in the pediatric age group.8585 Eken C, Serinken M, Dogan M. Ketamine may be related to reduced ejection fraction in children during the procedural sedation. Hum Exp Toxicol. 2016;36:106-10. The catecholaminergic effects mask myocardial depression. Further studies are needed to clarify the hemodynamic effects of this drug.

The post-sedation emergency phenomenon with ketamine, rare in pediatrics, can manifest itself through crying, lucid dreaming, hallucinations, and, more rarely, severe delirium. It is more common in adolescents when the drug is administered intramuscularly or at high doses.8686 Treston G, Bell A, Cardwell R, Fincher G, Chand D, Cashion G. What is the nature of the emergence phenomenon when using intravenous or intramuscular ketamine for paediatric procedural sedation?. Emerg Med Australas. 2009;21:315-22.,8787 Green SM, Roback MG, Krauss B, Brown L, McGlone RG, Agrawal D, et al. Predictors of emesis and recovery agitation with emergency department ketamine sedation: an individual-patient data meta-analysis of 8,282 children. Ann Emerg Med. 2009;54:171-80. There is no evidence that an infusion of midazolam as premedication to ketamine reduces the incidence of emergency phenomena. Wathen et al. found similar rates of emergence phenomena in pediatric patients receiving ketamine alone (7.1%) and in those receiving it in association with midazolam (6.2%).8888 Wathen JE, Roback MG, Mackenzie T, Bothner JP. Does midazolam alter the clinical effects of intravenous ketamine sedation in children? A double-blind, randomized, controlled, emergency department trial. Ann Emerg Med. 2000;36:579-88. Also, another study published by Sherwin et al. did not demonstrate any additional benefit of midazolam to prevent emergency phenomena.8989 Sherwin TS, Green SM, Khan A, Chapman DS, Dannenberg B. Does adjunctive midazolam reduce recovery agitation after ketamine sedation for pediatric procedures? A randomized, double-blind, placebo-controlled trial. Ann Emerg Med. 2000;35:229-38.

Ketamine is contraindicated in patients younger than 3 months due to the risk of airway complications, and in schizophrenic patients, due to the risk of psychotic stimulation.9090 Lahti AC, Koffel B, LaPorte D, Tamminga CA. Subanesthetic doses of ketamine stimulate psychosis in schizophrenia. Neuropsychopharmacology. 1995;13:9-19. Risk factors for adverse airway and breathing events are high intravenous doses, age less than 2 years or greater than 13 years, and co-administration of anticholinergics and benzodiazepines.9191 Green SM, Roback MG, Krauss B, Brown L, McGlone RG, Agrawal D, et al. Predictors of airway and respiratory adverse events with ketamine sedation in the emergency department: an individual-patient data meta-analysis of 8,282 children. Ann Emerg Med. 2009;54:158-68. The associated use of atropine may reduce hypersalivation, but does not reduce the frequency of adverse events.9292 Heinz P, Geelhoed GC, Wee C, Pascoe EM. Is atropine needed with ketamine sedation? A prospective, randomised, double blind study. Emerg Med J. 2006;23:206-9. Two randomized studies have demonstrated the reduction of hypersalivation with the prophylactic use of atropine, with no positive impact on the rate of adverse events.9393 Kye YC, Rhee JE, Kim K, Kim T, Jo YH, Jeong JH, et al. Clinical effects of adjunctive atropine during ketamine sedation in pediatric emergency patients. Am J Emerg Med. 2012;30:1981-5.,9494 Asadi P, Ghafouri HB, Yasinzadeh M, Kasnavieh SM, Modirian E. Ketamine and atropine for pediatric sedation: a prospective double-blind randomized controlled trial. Pediatr Emerg Care. 2013;29:136-9.

The use of ketamine should be avoided in patients with heart disease, with active pulmonary pathologies, or with central nervous system abnormalities. Currently, there is no contraindication to its use in traumatic brain injury, as there is no increase in the risk of intracranial hypertension or neurological complications, even in severe cases.9595 Bar-Joseph G, Guilburd Y, Tamir A, Guilburd JN. Effectiveness of ketamine in decreasing intracranial pressure in children with intracranial hypertension. J Neurosurg Pediatr. 2009;4:40-6.

96 Zeiler FA, Teitelbaum J, West M, Gillman LM. The ketamine effect on ICP in traumatic brain injury. Neurocrit Care. 2014;21:163-73.

97 Wang X, Ding X, Tong Y, Zong J, Zhao X, Ren H, et al. Ketamine does not increase intracranial pressure compared with opioids: meta-analysis of randomized controlled trials. J Anesth. 2014;28:821-7.
-9898 Zeiler FA, Teitelbaum J, West M, Gillman LM. The ketamine effect on intracranial pressure in nontraumatic neurological illness. J Crit Care. 2014;29:1096-106. A systematic review showed no significant difference in cerebral perfusion pressure, neurological outcome, length of stay in the intensive care unit (ICU), or mortality when the use of ketamine was compared with other sedatives.9999 Cohen L, Athaide V, Wickham ME, Doyle-Waters MM, Rose NG, Hohl CM. The effect of ketamine on intracranial and cerebral perfusion pressure and health outcomes: a systematic review. Ann Emerg Med. 2015;65:43-51. However, the relative contra-indication still remains in patients with intracranial masses, intracranial anatomical alterations, and hydrocephalus.100100 Green SM, Andolfatto G, Krauss BS. Ketamine and intracranial pressure: no contraindication except hydrocephalus. Ann Emerg Med. 2015;65:52-4.

Propofol

Propofol is a hypnotic sedative agent with fast and short-acting anesthetic properties, corresponding to 2,6-diisopropylphenol, which exerts its hypnotic action through the activation of GABA, a central inhibitory neurotransmitter.101101 Morgan DJ, Campbell GA, Crankshaw DP. Pharmacokinetics of propofol when given by intravenous infusion. Br J Clin Pharmacol. 1990;30:144-8.,102102 Marik PE. Propofol: therapeutic indications and side-effects. Curr Pharm Des. 2004;10:3639-49. It can be used in brief procedures, associated or unassociated with analgesic agents, such as fentanyl and ketamine.103103 Godambe SA, Elliot V, Matheny D, Pershad J. Comparison of propofol/fentanyl versus ketamine/midazolam for brief orthopedic procedural sedation in a pediatric emergency department. Pediatrics. 2003;112:116-23.,104104 Mallory MD, Baxter AL, Yanosky DJ, Cravero JP. Pediatric Sedation Research Consortium. Emergency physician-administered propofol sedation: a report on 25,433 sedations from the pediatric sedation research consortium. Ann Emerg Med. 2011;57:462-8.

For longer procedures, such as in MRI sedation, propofol can be used under continuous infusion.105105 Srinivasan M, Turmelle M, Depalma LM, Mao J, Carlson DW. Procedural sedation for diagnostic imaging in children by pediatric hospitalists using propofol: analysis of the nature, frequency, and predictors of adverse events and interventions. J Pediatr. 2012;160:801-6. Sebe et al. published a prospective pediatric study with a cohort of 200 pediatric patients undergoing imaging examinations, in which propofol was more effective than midazolam in terms of sedation effectiveness and shorter recovery time, with no statistical difference in the rate of adverse events between the drugs.106106 Sebe A, Yilmaz HL, Koseoglu Z, Ay MO, Gulen M. Comparison of midazolam and propofol for sedation in pediatric diagnostic imaging studies. Postgrad Med. 2014;126:225-30. Studies comparing propofol, pentobarbital, and dexmedetomidine in MRI sedation were also favorable to the first drug.107107 Mallory MD, Baxter AL, Kost SI. Pediatric Sedation Research Consortium. Propofol vs pentobarbital for sedation of children undergoing magnetic resonance imaging: results from the Pediatric Sedation Research Consortium. Paediatr Anaesth. 2009;19:601-11.

108 Pershad J, Wan J, Anghelescu DL. Comparison of propofol with pentobarbital/midazolam/fentanyl sedation for magnetic resonance imaging of the brain in children. Pediatrics. 2007;120:e629-36.
-109109 Fang H, Yang L, Wang X, Zhu H. Clinical efficacy of dexmedetomidine versus propofol in children undergoing magnetic resonance imaging: a meta-analysis. Int J Clin Exp Med. 2015;8:11881-9. More complex and time-consuming imaging studies can be performed effectively with continuous infusion, without increasing recovery time or risk of adverse effects.110110 Griffiths MA, Kamat PP, McCracken CE, Simon HK. Is procedural sedation with propofol acceptable for complex imaging? A comparison of short vs. prolonged sedations in children. Pediatr Radiol. 2013;43:1273-8.

The metabolism of propofol is hepatic, the elimination is biphasic (with the initial phase at 40 min and terminal phase at 4-7 h), and excretion is mostly urinary (88%). Administration is exclusively IV, usually associated with pain during infusion, an effect that can be reduced with pre-treatment of the vein with lidocaine, i.e., previous administration of small doses of opioid or ketamine or infusion into large-caliber veins.111111 Jalota L, Kalira V, George E, Shi YY, Hornuss C, Radke O, et al. Prevention of pain on injection of propofol: systematic review and meta-analysis. BMJ. 2011;342:d1110.

Propofol has several cardiovascular effects, with hypotension being the most significant.112112 Chidambaran V, Costandi A, D'Mello A. Propofol: a review of its role in pediatric anesthesia and sedation. CNS Drugs. 2015;29:543-63. This is due to arterial vasodilation through the reduction of sympathetic tonus, but also because it affects myocardial contractility and cardiac output.113113 Robinson BJ, Ebert TJ, O'Brien TJ, Colinco MD, Muzi M. Mechanisms whereby propofol mediates peripheral vasodilation in humans. Sympathoinhibition or direct vascular relaxation?. Anesthesiology. 1997;86:64-72. These effects may be exacerbated in hypovolemic patients, or patients with pre-existing heart disease or the concomitant use of other cardiac depressant drugs. Bolus administration of saline 0.9% at 20 mL/kg prior to propofol infusion did not reduce the risk of hypotension in pediatric patients when compared to the control group in a randomized study published by Jager et al.114114 Jager MD, Aldag JC, Deshpande GG. A presedation fluid bolus does not decrease the incidence of propofol-induced hypotension in pediatric patients. Hosp Pediatr. 2015;5:85-91. The decrease in the systemic vascular resistance induced by the drug may be harmful in patients with congenital heart disease; however, the heart rate does not change significantly. Apnea and airway obstruction is frequent, even at usual doses, since propofol depresses the CNS, which can reduce respiratory rate and pulmonary volume.115115 Marik PE. Propofol: therapeutic indications and side effects. Curr Pharm Des. 2004;10:3639-49. The risk of respiratory depression increases proportionally to the infusion velocity as well as the risk of bradycardia, and hypotension is associated with higher doses.116116 Dosani M, McCormack J, Reimer E, Brant R, Dumont G, Lim J, et al. Slower administration of propofol preserves adequate respiration in children. Paediatr Anaesth. 2010;20:1001-8.,117117 Milius EM, Papademetrious TR, Heitlinger LA. Retrospective review of propofol dosing for procedural sedation in pediatric patients. J Pediatr Pharmacol Ther. 2012;17:246-51. It is contraindicated in patients with allergy to eggs, soybeans, and their derivatives.118118 Murphy A, Campbell DE, Baines D, Mehr S. Allergic reactions to propofol in egg-allergic children. Anesth Analg. 2011;113:140-4.

Despite the adverse effects, propofol is safe and effective when performed with adequate monitoring. Mallory et al., in a retrospective study that analyzed 25,433 episodes of sedation with propofol, showed a severe adverse event rate, such as airway obstruction, apnea, and oxygen saturation drop in 2.2%, with no associated death reports related to this drug.104104 Mallory MD, Baxter AL, Yanosky DJ, Cravero JP. Pediatric Sedation Research Consortium. Emergency physician-administered propofol sedation: a report on 25,433 sedations from the pediatric sedation research consortium. Ann Emerg Med. 2011;57:462-8. Chiaretti et al. reviewed 36,156 procedures and obtained an even lower complication rate of 0.75%.119119 Chiaretti A, Benini F, Pierri F, Vecchiato K, Ronfani L, Agosto C, et al. Safety and efficacy of propofol administered by paediatricians during procedural sedation in children. Acta Paediatr. 2014;103:182-7.

Propofol infusion syndrome is defined as refractory acute bradycardia progressing to asystole, metabolic acidosis, rhabdomyolysis, hyperlipidemia, and hepatic steatosis. It is described in severely-ill pediatric patients receiving this medication for a prolonged period of time (>48 h) and at high doses (>4 mcg/kg/min).120120 Vasile B, Rasulo F, Candiani A, Latronico N. The pathophysiology of propofol infusion syndrome: a simple name for a complex syndrome. Intensive Care Med. 2003;29:1417-25.

Ketofol

The use of ketamine and propofol in association, known as “ketofol,” has become popular due to the possibility of counterbalancing the side effects of each medication and enhanced sedation, increasing efficacy and safety.121121 Alletag MJ, Auerbach MA, Baum CR. Ketamine, propofol, and ketofol use for pediatric sedation. Pediatr Emerg Care. 2012;28:1391-5; quiz 1396-8. Ketamine maintains the respiratory drive, prevents hypotension and bradycardia, and provides analgesia, while propofol reduces the incidence of nausea and vomiting, in addition to hypothetically prevents emergency reactions.122122 Andolfatto G, Willman E. A prospective case series of pediatric procedural sedation and analgesia in the emergency department using single-syringe ketamine-propofol combination (ketofol). Acad Emerg Med. 2010;17:194-201.

When compared to propofol as a single drug, the use of ketofol in pediatric patients resulted in fewer respiratory adverse events, hypotension, and bradycardia, as well as fewer additional doses of the drug.123123 David H, Shipp J. A randomized controlled trial of ketamine/propofol versus propofol alone for emergency department procedural sedation. Ann Emerg Med. 2011;57:435-41. A meta-analysis of 932 patients showed fewer respiratory adverse events with the ketofol group compared to propofol (29% vs. 35.4%), with no significant difference in the proportion of general adverse events (38.8% vs. 42.5%).124124 Yan JW, McLeod SL, Iansavitchene A. Ketamine-propofol versus propofol alone for procedural sedation in the emergency department: a systematic review and meta-analysis. Acad Emerg Med. 2015;22:1003-13. Another meta-analysis by Jalili et al. demonstrated that ketofol significantly reduced respiratory and cardiovascular complications (hypotension and bradycardia).125125 Jalili M, Bahreini M, Doosti-Irani A, Masoomi R, Arbab M, Mirfazaelian H. Ketamine-propofol combination (ketofol) vs propofol for procedural sedation and analgesia: systematic review and meta-analysis. Am J Emerg Med. 2016;34:558-69.

There is no consensus regarding which dose of each drug should be used when in combination, with the suggestion of an initial dose of 0.5 mg/kg of propofol and 1 mg/kg of ketamine. No difference in the quality of sedation or safety profile was observed between mixtures at the proportions of 1:1 and 1:4 of ketamine and propofol in adults.126126 Miner JR, Moore JC, Austad EJ, Plummer D, Hubbard L, Gray RO. Randomized, double-blinded, clinical trial of propofol, 1:1 propofol/ketamine, and 4:1 propofol/ketamine for deep procedural sedation in the emergency department. Ann Emerg Med. 2015;65:479-88. Further studies on the use of ketofol in pediatric patients are required to elucidate its advantages and disadvantages as a sedative agent.127127 Green SM, Andolfatto G, Krauss BS. Ketofol for procedural sedation revisited: pro and con. Ann Emerg Med. 2015;65:489-91.

Dexmedetomidine

Dexmedetomidine (DEX) is an alpha-2 adrenergic agonist with an action that is not mediated by GABA, which promotes sedation without decreasing the respiratory drive.128128 Belleville JP, Ward DS, Bloor BC, Maze M. Effects of intravenous dexmedetomidine in humans. I. Sedation, ventilation, and metabolic rate. Anesthesiology. 1992;77:1125-33. Despite its off-label use in pediatrics, its use has been increasingly used for short procedures and also for situations requiring prolonged sedation.129129 Tobias JD. Dexmedetomidine: applications in pediatric critical care and pediatric anesthesiology. Pediatr Crit Care Med. 2007;8:115-31. It can be used as a single agent or in combination with midazolam, ketamine, or opioids.130130 Mahmoud M, Mason KP. Dexmedetomidine: review, update, and future considerations of paediatric perioperative and periprocedural applications and limitations. Br J Anaesth. 2015;:115171-82.,131131 Mason KP, Lerman J. Review article: dexmedetomidine in children: current knowledge and future applications. Anesth Analg. 2011;113:1129-42. In the emergency department, it is mainly used for imaging studies.132132 Mason KP, Lubisch NB, Robinson F, Roskos R. Intramuscular dexmedetomidine sedation for pediatric MRI and CT. AJR Am J Roentgenol. 2011;197:720-5.,133133 Mekitarian Filho E, Robinson F, de Carvalho WB, Gilio AE, Mason KP. Intranasal dexmedetomidine for sedation for pediatric computed tomography imaging. J Pediatr. 2015;166:1313-5. Due to its unique pharmacological characteristics, DEX is used to induce an electroencephalogram (EEG) pattern similar to that of natural sleep.134134 Mason KP, O'Mahony E, Zurakowski D, Libenson MH. Effects of dexmedetomidine sedation on the EEG in children. Paediatr Anaesth. 2009;19:1175-83.,135135 Baier NM, Mendez SS, Kimm D, Velazquez AE, Schroeder AR. Intranasal dexmedetomidine: an effective sedative agent for electroencephalogram and auditory brain response testing. Paediatr Anaesth. 2016;26:280-5. It is an effective alternative to midazolam in sedation during upper digestive endoscopy, with better sedation potential and fewer adverse effects.136136 Zhang F, Sun HR, Zheng ZB, Liao R, Liu J. Dexmedetomidine versus midazolam for sedation during endoscopy: a meta-analysis. Exp Ther Med. 2016;11:2519-24.

It has hepatic metabolism and urinary excretion (95%), with IV, IN, oral, and oral gastric mucosa routes, with the latter showing less bioavailability.128128 Belleville JP, Ward DS, Bloor BC, Maze M. Effects of intravenous dexmedetomidine in humans. I. Sedation, ventilation, and metabolic rate. Anesthesiology. 1992;77:1125-33. It can cause hypotension, bradycardia, and sinus arrhythmia.130130 Mahmoud M, Mason KP. Dexmedetomidine: review, update, and future considerations of paediatric perioperative and periprocedural applications and limitations. Br J Anaesth. 2015;:115171-82.,137137 Carney L, Kendrick J, Carr R. Safety and effectiveness of dexmedetomidine in the pediatric intensive care unit (SAD-PICU). Can J Hosp Pharm. 2013;66:21-7. Due to its pharmacological characteristics and its clinical applications, dexmedetomidine appears to be a good alternative to the use of chloral hydrate in pediatric emergency departments after the latter's recent discontinuation of use in Brazil.

Although it has not been approved by the Food and Drug Administration for pediatric use due to lack of data demonstrating its safety profile, studies have already shown that it is a safe sedative, especially in the context of adult and pediatric ICUs.138138 Department of Health and Human Services. Public Health Service. Food and Drug Administration. Center for Drug Evaluation and Research. Office of Surveillance and Epidemiology. Pediatric postmarketing pharmacovigilance and drug utilization review. Precedex (dexmedetomidine HCl injection); 2016 Mar. Available from: https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/PediatricAdvisoryCommittee/UCM494470.pdf [cited 18.11.16].
https://www.fda.gov/downloads/AdvisoryCo...
Constantin et al. published a meta-analysis of 1994 adult ICU patients, showing a reduction in hospitalization time, mechanical ventilation time, and delirium occurrence in 48 h, but an increase in cases of bradycardia and hypotension was observed.139139 Constantin JM, Momon A, Mantz J, Payen JF, De Jonghe B, Perbet S, et al. Efficacy and safety of sedation with dexmedetomidine in critical care patients: a meta-analysis of randomized controlled trials. Anaesth Crit Care Pain Med. 2016;35:7-15. Berkenbosch et al., in a prospective pediatric study, demonstrated the efficacy and safety of dexmedetomidine for noninvasive procedures.140140 Berkenbosch JW, Wankum PC, Tobias JD. Prospective evaluation of dexmedetomidine for noninvasive procedural sedation in children. Pediatr Crit Care Med. 2005;6:435-9, quiz 440.

Etomidate

Etomidate is an imidazole derivative used as an ultra-fast acting sedative-hypnotic agent that binds to GABA receptors in the central nervous system. Commonly used in rapid intubation sequences in children, it may be used in non-painful short-term procedures, such as skull CT, and in painful procedures associated with an analgesic drug. It has few hemodynamic repercussions, and respiratory effects are rare.141141 Forman SA. Clinical and molecular pharmacology of etomidate. Anesthesiology. 2011;114:695-707.,142142 Falk J, Zed PJ. Etomidate for procedural sedation in the emergency department. Ann Pharmacother. 2004;38:1272-7.

It is a highly lipophilic drug, with hepatic metabolism and urinary excretion (75%). It is administered exclusively intravenously, and may be irritating at the infusion site; it is preferable to administer it through larger caliber veins or in combination with lidocaine.141141 Forman SA. Clinical and molecular pharmacology of etomidate. Anesthesiology. 2011;114:695-707. It can cause myoclonus (without EEG repercussions), nausea, vomiting, apnea, and hypoventilation.142142 Falk J, Zed PJ. Etomidate for procedural sedation in the emergency department. Ann Pharmacother. 2004;38:1272-7.,143143 Mandt MJ, Roback MG, Bajaj L, Galinkin JL, Gao D, Wathen JE. Etomidate for short pediatric procedures in the emergency department. Pediatr Emerg Care. 2012;28:898-904. Di Liddo et al. compared etomidate and midazolam in the sedation of pediatric patients submitted to fracture reduction and showed a higher proportion of adequately sedated patients in the etomidate group (92% vs. 36%), with a shorter time of induction and recovery.144144 Di Liddo L, D'Angelo A, Nguyen B, Bailey B, Amre D, Stanciu C. Etomidate versus midazolam for procedural sedation in pediatric outpatients: a randomized controlled trial. Ann Emerg Med. 2006;48:433-40, 440.e1.

As etomidate inhibits the 11-beta-hydroxylase enzyme, which participates in the production of adrenal hormones, a single dose can suppress the stress-induced cortisol production response and may last 6-8 h.145145 Majesko A, Darby JM. Etomidate and adrenal insufficiency: the controversy continues. Crit Care. 2010;14:338.,146146 Wagner RL, White PF, Kan PB, Rosenthal MH, Feldman D. Inhibition of adrenal steroidogenesis by the anesthetic etomidate. N Engl J Med. 1984;310:1415-21. Thus, the use of this drug in septic or critically-ill patients, even in a single dose, remains controversial.145145 Majesko A, Darby JM. Etomidate and adrenal insufficiency: the controversy continues. Crit Care. 2010;14:338. Nonetheless, its use in healthy patients appears to be well tolerated.147147 Jones AE. The etomidate debate. Ann Emerg Med. 2010;56:490-1. Two prospective randomized studies in adults did not demonstrate increased morbidity and mortality or hospital length of stay when etomidate was administered by bolus in the rapid intubation sequence.148148 Tekwani KL, Watts HF, Sweis RT, Rzechula KH, Kulstad EB. A comparison of the effects of etomidate and midazolam on hospital length of stay in patients with suspected sepsis: a prospective, randomized study. Ann Emerg Med. 2010;56:481-9.

Conclusion

Painful diagnostic and therapeutic procedures that frequently require patient collaboration are common in the practice of pediatric emergency. Indication of analgesia and sedation for such procedures should occur after a careful patient assessment, considering the purpose, risks, and benefits associated with the procedure and the use of medications. The use of protocols for this purpose should guide the professional in the choice of medication, the appropriate material, and evaluation of the discharge criteria, thus ensuring quality in care. Analgesia and sedation for procedures requires training of health teams, aiming at safety and effectiveness. Studies have shown the best pharmacological choices for certain procedures, considering the age and health status of each patient, making sedoanalgesia an increasingly common and safer practice in the pediatric emergency room.

  • Please cite this article as: Ramalho CE, Bretas PM, Schvartsman C, Reis AG. Sedation and analgesia for procedures in the pediatric emergency room. J Pediatr (Rio J). 2017;93:2-18.
  • ☆☆
    Study carried out at Universidade de São Paulo (USP), Faculdade de Medicina, Hospital das Clínicas, Instituto da Criança, São Paulo, SP, Brazil.

References

  • 1
    Krauss B, Green SM. Procedural sedation and analgesia in children. Lancet. 2006;367:766-80.
  • 2
    McCoy S, Lyttle MD, Hartshorn S, Larkin P, Brenner M, O'Sullivan R, et al. A qualitative study of the barriers to procedural sedation practices in paediatric emergency medicine in the UK and Ireland. Emerg Med J. 2016;33:527-32.
  • 3
    Mason KP. Challenges in paediatric procedural sedation: political, economic, and clinical aspects. Br J Anaesth. 2014;113, Sii48-62.
  • 4
    Gozal D, Mason KP. Pediatric sedation: a global challenge. Int J Pediatr. 2010;:701257.
  • 5
    Bartolome SM, Cid JL, Freddi N. Analgesia and sedation in children: practical approach for the most frequent situations. J Pediatr (Rio J). 2007;83:S71-82.
  • 6
    Miyake RS, Reis AG, Grisi S. Sedation and analgesia for children. Rev Assoc Med Bras. 1998;44:56-64.
  • 7
    Carvalho WB, Troster EJ. Sedation and analgesia at the emergency room. J Pediatr (Rio J). 1999;75:S294-306.
  • 8
    Manyande A, Cyna AM, Yip P, Chooi C, Middleton P. Non-pharmacological interventions for assisting the induction of anaesthesia in children. Cochrane Database Syst Rev. 2015;14:CD006447.
  • 9
    Birnie KA, Noel M, Parker JA, Chambers CT, Uman LS, Kisely SR, et al. Systematic review and meta-analysis of distraction and hypnosis for needle-related pain and distress in children and adolescents. J Pediatr Psychol. 2014;39:783-808.
  • 10
    White PF, Way WL, Trevor AJ. Ketamine - its pharmacology and therapeutic uses. Anesthesiology. 1982;56:119-36.
  • 11
    Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology. 2002;96:1004-17.
  • 12
    Büttner W, Finke W. Analysis of behavioural and physiological parameters for the assessment of postoperative analgesic demand in newborns, infants and young children: a comprehensive report on seven consecutive studies. Paediatr Anaesth. 2000;10:303-18.
  • 13
    Gonsalves S, Mercer J. Physiological correlates of painful stimulation in preterm infants. Clin J Pain. 1993;9:88-93.
  • 14
    Crellin D, Sullivan TP, Babl FE, O'Sullivan R, Hutchinson A. Analysis of the validation of existing behavioral pain and distress scales for use in the procedural setting. Paediatr Anaesth. 2007;17:720-33.
  • 15
    Lawrence J, Alcock D, McGrath P, Kay J, MacMurray SB, Dulberg C. The development of a tool to assess neonatal pain. Neonatal Netw. 1993;12:59-66.
  • 16
    Merkel SI, Voepel-Lewis T, Shayevitz JR, Malviya S. The FLACC: a behavioral scale for scoring postoperative pain in young children. Pediatr Nurs. 1997;23:293-7.
  • 17
    Manworren RC, Hynan LS. Clinical validation of FLACC: preverbal patient pain scale. Pediatr Nurs. 2003;29:140-6.
  • 18
    Beyer JE, Aradine CR. Content validity of an instrument to measure young children's perceptions of the intensity of their pain. J Pediatr Nurs. 1986;1:386-95.
  • 19
    Bieri D, Reeve RA, Champion GD, Addicoat L, Ziegler JB. The Faces Pain Scale for the self-assessment of the severity of pain experienced by children: development, initial validation, and preliminary investigation for ratio scale properties. Pain. 1990;41:139-50.
  • 20
    Garra G, Singer AJ, Taira BR, Chohan J, Cardoz H, Chisena E, et al. Validation of the Wong-Baker FACES Pain Rating Scale in pediatric emergency department patients. Acad Emerg Med. 2010;17:50-4.
  • 21
    Todd KH, Funk KG, Funk JP, Bonacci R. Clinical significance of reported changes in pain severity. Ann Emerg Med. 1996;27:485-9.
  • 22
    Tobias JD, Leder M. Procedural sedation: a review of sedative agents, monitoring, and management of complications. Saudi J Anaesth. 2011;5:395-410.
  • 23
    Bellolio MF, Puls HA, Anderson JL, Gilani WI, Murad MH, Barrionuevo P, et al. Incidence of adverse events in paediatric procedural sedation in the emergency department: a systematic review and meta-analysis. BMJ Open. 2016;6:e011384.
  • 24
    Cravero JP, Blike GT, Beach M, Gallagher SM, Hertzog JH, Havidich JE, et al. Incidence and nature of adverse events during pediatric sedation/anesthesia for procedures outside the operating room: report from the Pediatric Sedation Research Consortium. Pediatrics. 2006;118:1087-96.
  • 25
    Bellolio MF, Gilani WI, Barrionuevo P, Murad MH, Erwin PJ, Anderson JR, et al. Incidence of adverse events in adults undergoing procedural sedation in the emergency department: a systematic review and meta-analysis. Acad Emerg Med. 2016;23:119-34.
  • 26
    American Society of Anesthesiologists Committee. Practice guidelines for preoperative fasting and the use of pharmacologic agents to reduce the risk of pulmonary aspiration: application to healthy patients undergoing elective procedures: an updated report by the American Society of Anesthesiologists Committee on Standards and Practice Parameters. Anesthesiology. 2011;114:495-511.
  • 27
    Godwin SA, Burton JH, Gerardo CJ, Hatten BW, Mace SE, Silvers SM, et al. Clinical policy: procedural sedation and analgesia in the emergency department. Ann Emerg Med. 2014;63:247-258.e18.
  • 28
    Thorpe RJ, Benger J. Pre-procedural fasting in emergency sedation. Emerg Med J. 2010;27:254-61.
  • 29
    Hoffman GM, Nowakowski R, Troshynski TJ, Berens RJ, Weisman SJ. Risk reduction in pediatric procedural sedation by application of an American Academy of Pediatrics/American Society of Anesthesiologists process model. Pediatrics. 2002;109:236-43.
  • 30
    Clark M, Birisci E, Anderson JE, Anliker CM, Bryant MA, Downs C, et al. The risk of shorter fasting time for pediatric deep sedation. Anesth Essays Res. 2016;10:607-12.
  • 31
    Butler MG, Hayes BG, Hathaway MM, Begleiter ML. Specific genetic diseases at risk for sedation/anesthesia complications. Anesth Analg. 2000;91:837-55.
  • 32
    Malviya S, Voepel-Lewis T, Tait AR. Adverse events and risk factors associated with the sedation of children by nonanesthesiologists. Anesth Analg. 1997;85:1207-13.
  • 33
    Jirativanont T, Manomayangkul K, Udomphorn Y, Yokubol B, Saguansab A, Kraiprasit K, et al. Incidence and risk factors for adverse events during anesthesiologist-led sedation or anesthesia for diagnostic imaging in children: a prospective, observational cohort study. Asi Biomed. 2017;9:649-58.
  • 34
    Caperell K, Pitetti R. Is higher ASA class associated with an increased incidence of adverse events during procedural sedation in a pediatric emergency department?. Pediatr Emerg Care. 2009;25:661-4.
  • 35
    Nargozian C. The airway in patients with craniofacial abnormalities. Paediatr Anaesth. 2004;14:53-9.
  • 36
    Heinrich S, Birkholz T, Ihmsen H, Irouschek A, Ackermann A, Schmidt J. Incidence and predictors of difficult laryngoscopy in 11,219 pediatric anesthesia procedures. Paediatr Anaesth. 2012;22:729-36.
  • 37
    Heinrich S, Birkholz T, Irouschek A, Ackermann A, Schmidt J. Incidences and predictors of difficult laryngoscopy in adult patients undergoing general anesthesia: a single-center analysis of 102,305 cases. J Anesth. 2013;27:815-21.
  • 38
    Coté CJ, Wilson S. Guidelines for monitoring and management of pediatric patients before, during, and after sedation for diagnostic and therapeutic procedures: update 2016. Pediatrics. 2016;138:pii: e20161212.
  • 39
    Waugh JB, Epps CA, Khodneva YA. Capnography enhances surveillance of respiratory events during procedural sedation: a meta-analysis. J Clin Anesth. 2011;23:189-96.
  • 40
    Langhan ML, Shabanova V, Li FY, Bernstein SL, Shapiro ED. A randomized controlled trial of capnography during sedation in a pediatric emergency setting. Am J Emerg Med. 2015;33:25-30.
  • 41
    Krauss BS, Krauss BA, Green SM. Procedural sedation and analgesia in children. N Engl J Med. 2014;370:e23.
  • 42
    Couloures KG, Beach M, Cravero JP, Monroe KK, Hertzog JH. Impact of provider specialty on pediatric procedural sedation complication rates. Pediatrics. 2011;127:e1154-60.
  • 43
    Sauter TC, Hautz WE, Hostettler S, Brodmann-Maeder M, Martinolli L, Lehmann B, et al. Interprofessional and interdisciplinary simulation-based training leads to safe sedation procedures in the emergency department. Scand J Trauma Resusc Emerg Med. 2016;24:97.
  • 44
    Farnsworth ST, Egan TD, Johnson SE, Westenskow D. Teaching sedation and analgesia with simulation. J Clin Monit Comput. 2000;16:273-85.
  • 45
    Fehr JJ, Chao J, Kuan C, Zhong J. The important role of simulation in sedation. Curr Opin Anaesthesiol. 2016;29:S14-20.
  • 46
    Krauss B, Green SM. Training and credentialing in procedural sedation and analgesia in children: lessons from the United States model. Paediatr Anaesth. 2008;18:30-5.
  • 47
    Zaveri PP, Davis AB, O'Connell KJ, Willner E, Schinasi DA, Ottolini M. Virtual reality for pediatric sedation: a randomized controlled trial using simulation. Cureus. 2016;8:e486.
  • 48
    Maghraby N, Pearson E, Xue X, Colacone A, Afilalo M. What is the impact of the implementation of an evidence based procedural sedation protocol in the emergency department?. J Clin Trials. 2016;6:283.
  • 49
    Blumer JL. Clinical pharmacology of midazolam in infants and children. Clin Pharmacokinet. 1998;35:37-47.
  • 50
    Wright SW, Chudnofsky CR, Dronen SC, Kothari R, Birrer P, Blanton DM, et al. Comparison of midazolam and diazepam for conscious sedation in the emergency department. Ann Emerg Med. 1993;22:201-5.
  • 51
    Ilkhanipour K, Juels CR, Langdorj MI. Pediatric pain control and conscious sedation: a survey of emergency medicine residencies. Acad Emerg Med. 1994;1:368-72.
  • 52
    Sukys GA, Schvartsman C, Reis AG. Evaluation of rapid sequence intubation in the pediatric emergency department. J Pediatr (Rio J). 2011;87:343-9.
  • 53
    Notterman DA. Sedation with intravenous midazolam in the pediatric intensive care unit. Clin Pediatr (Phila). 1997;36:449-54.
  • 54
    Griffith N, Howell S, Mason DG. Intranasal midazolam for premedication of children undergoing day-case anesthesia: comparison of two delivery systems with assessment of intra-observer variability. Br J Anesth. 1998;81:865-9.
  • 55
    Sahyoun C, Kraus B. Clinical implications of pharmacokinetics and pharmacodynamics of procedural sedation agents in children. Curr Opin Pediatr. 2012;24:225-32.
  • 56
    Smith D, Cheek H, Denson B, Pruitt CM. Lidocaine pretreatment reduces the discomfort of intranasal midazolam administration: a randomized, double-blind, placebo-controlled trial. Acad Emerg Med. 2017;24:161-7.
  • 57
    Chiaretti A, Barone G, Rigante D, Ruggiero A, Pierri F, Barbi E, et al. Intranasal lidocaine and midazolam for procedural sedation in children. Arch Dis Child. 2011;96:160-3.
  • 58
    Lucas SS, Nasr VG, Ng AJ, Joe C, Bond M, DiNardo JA. Pediatric Cardiac Intensive Care Society 2014 consensus statement: pharmacotherapies in cardiac critical care: sedation, analgesia and muscle relaxant. Pediatr Crit Care Med. 2016;17:S3-15.
  • 59
    Shannon M, Albers G, Burkart K, Liebelt E, Kelley M, McCubbin MM, et al. Safety and efficacy of flumazenil in the reversal of benzodiazepine-induced conscious sedation. J Pediatr. 1997;131:582-6.
  • 60
    Inturrisi CE. Clinical pharmacology of opioids for pain. Clin J Pain. 2002;18:S3-13.
  • 61
    Stanski DR, Greenblatt DJ, Lowenstein E. Kinetics of intravenous and intramuscular morphine. Clin Pharmacol Ther. 1978;24:52-9.
  • 62
    Barcelos A, Garcia PC, Portela JL, Piva JP, Garcia JP, Santana JC. Comparison of two analgesia protocols for the treatment of pediatric orthopedic emergencies. Rev Assoc Med Bras. 2015;61:362-7.
  • 63
    Gupta M, Msambichaka L, Ballas SK, Gupta K. Morphine for the treatment of pain in sickle cell disease. Sci World J. 2015;2015:540154.
  • 64
    Martin LD, Jimenez N, Lynn AM. A review of perioperative anesthesia and analgesia for infants: updates and trends to watch. F1000Res. 2017;6:120.
  • 65
    McRorie TI, Lynn AM, Nespeca MK, Opheim KE, Slattery JT. The maturation of morphine clearance and metabolism. Am J Dis Child. 1992;146:972-6.
  • 66
    Lynn A, Nespeca MK, Bratton SL, Strauss SG, Shen DD. Clearance of morphine in postoperative infants during intravenous infusion: the influence of age and surgery. Anesth Analg. 1998;86:958-63.
  • 67
    Rang HP, Dale MM, Ritter JM, Flower RJ, Henderson G. Farmacologia. 7th ed. Rio de Janeiro: Elsevier; 2012.
  • 68
    Tobias JD. Tolerance, withdrawal, and physical dependency after long-term sedation and analgesia of children in the pediatric intensive care unit. Crit Care Med. 2000;28:2122-32.
  • 69
    Feierman DE, Lasker JM. Metabolism of fentanyl, a synthetic opioid analgesic, by human liver microsomes: role of CYP3A4. Drug Metab Dispos. 1996;24:932-9.
  • 70
    Labroo RB, Paine MF, Thummel KE, Kharasch ED. Fentanyl metabolism by human hepatic and intestinal cytochrome P4503A4: implications for interindividual variability in disposition, efficacy, and drug interactions. Drug Metab Dispos. 1997;25:1072-80.
  • 71
    Singleton MA, Rosen JI, Fisher DM. Plasma concentrations of fentanyl in infants, children and adults. Can J Anaesth. 1987;34:152-5.
  • 72
    Lexi-Comp Inc. Lexicomp Online, Pediatric and Neonatal Lexi-Drugs Online. Hudson, OH: Lexi-Comp, Inc, April-May 2017; 2017.
  • 73
    Miner JR, Kletti C, Herold M, Hubbard D, Biros MH. Randomized clinical trial of nebulized fentanyl citrate versus i.v. fentanyl citrate in children presenting to the emergency department with acute pain. Acad Emerg Med. 2007;14:895-8.
  • 74
    Borland M, Milsom S, Esson A. Equivalency of two concentrations of fentanyl administered by the intranasal route for acute analgesia in children in a paediatric emergency department: a randomized controlled trial. Emerg Med Aust. 2011;23:202-8.
  • 75
    Young PA, Nicol MF, Kendall JM, Harrington AP. A prospective randomized pilot comparison of intranasal fentanyl and intramuscular morphine for analgesia in children presenting to the emergency department with clinical fractures. Emerg Med. 1999;11:90-4.
  • 76
    Roback MG, Wathen JE, Bajaj L, Bothner JP. Adverse events associated with procedural sedation and analgesia in a pediatric emergency department: a comparison of common parenteral drugs. Acad Emerg Med. 2005;12:508-13.
  • 77
    Fleischman RJ, Frazer DG, Daya MJ, Jui J, Newgard CD. Effectiveness and safety of fentanyl compared with morphine for out-of-hospital analgesia. Prehosp Emerg Care. 2010;14:167-75.
  • 78
    Barsan WG, Seger D, Danzl DF, Ling LJ, Bartlett R, Buncher R, et al. Duration of antagonistic effects of nalmefene and naloxone in opiate-induced sedation for emergency department procedures. Am J Emerg. 1989;7:155-61.
  • 79
    Barrons RW, Woods JA. Low-dose naloxone for prophylaxis of postoperative nausea and vomiting: a systematic review and meta-analysis. Pharmacotherapy. 2017;37:546-54.
  • 80
    Green SM, Roback MG, Kennedy RM, Krauss B. Clinical practice guideline for emergency department ketamine dissociative sedation: 2011 update. Ann Emerg Med. 2011;57:449-61.
  • 81
    Deasy C, Babl FE. Intravenous vs intramuscular ketamine for pediatric procedural sedation by emergency medicine specialists: a review. Paediatr Anaesth. 2010;20:787-96.
  • 82
    Green SM, Roback MG, Krauss B, Brown L, McGlone RG, Agrawal D, et al. Emergency Department Ketamine Meta-Analysis Study Group. Predictors of emesis and recovery agitation with emergency department ketamine sedation: an individual-patient data meta-analysis of 8,282 children. Ann Emerg Med. 2009;54:171-80.
  • 83
    Kannikeswaran N, Lieh-Lai M, Malian M, Wang B, Farooqi A, Roback MG. Optimal dosing of intravenous ketamine for procedural sedation in children in the ED - a randomized controlled trial. Am J Emerg Med. 2016;34:1347-53.
  • 84
    Jakobsen CJ, Torp P, Vester AE. Ketamine reduce left ventricular systolic and diastolic function in patients with ischaemic heart disease. Acta Anaesthesiol Scand. 2010;54:1137-44.
  • 85
    Eken C, Serinken M, Dogan M. Ketamine may be related to reduced ejection fraction in children during the procedural sedation. Hum Exp Toxicol. 2016;36:106-10.
  • 86
    Treston G, Bell A, Cardwell R, Fincher G, Chand D, Cashion G. What is the nature of the emergence phenomenon when using intravenous or intramuscular ketamine for paediatric procedural sedation?. Emerg Med Australas. 2009;21:315-22.
  • 87
    Green SM, Roback MG, Krauss B, Brown L, McGlone RG, Agrawal D, et al. Predictors of emesis and recovery agitation with emergency department ketamine sedation: an individual-patient data meta-analysis of 8,282 children. Ann Emerg Med. 2009;54:171-80.
  • 88
    Wathen JE, Roback MG, Mackenzie T, Bothner JP. Does midazolam alter the clinical effects of intravenous ketamine sedation in children? A double-blind, randomized, controlled, emergency department trial. Ann Emerg Med. 2000;36:579-88.
  • 89
    Sherwin TS, Green SM, Khan A, Chapman DS, Dannenberg B. Does adjunctive midazolam reduce recovery agitation after ketamine sedation for pediatric procedures? A randomized, double-blind, placebo-controlled trial. Ann Emerg Med. 2000;35:229-38.
  • 90
    Lahti AC, Koffel B, LaPorte D, Tamminga CA. Subanesthetic doses of ketamine stimulate psychosis in schizophrenia. Neuropsychopharmacology. 1995;13:9-19.
  • 91
    Green SM, Roback MG, Krauss B, Brown L, McGlone RG, Agrawal D, et al. Predictors of airway and respiratory adverse events with ketamine sedation in the emergency department: an individual-patient data meta-analysis of 8,282 children. Ann Emerg Med. 2009;54:158-68.
  • 92
    Heinz P, Geelhoed GC, Wee C, Pascoe EM. Is atropine needed with ketamine sedation? A prospective, randomised, double blind study. Emerg Med J. 2006;23:206-9.
  • 93
    Kye YC, Rhee JE, Kim K, Kim T, Jo YH, Jeong JH, et al. Clinical effects of adjunctive atropine during ketamine sedation in pediatric emergency patients. Am J Emerg Med. 2012;30:1981-5.
  • 94
    Asadi P, Ghafouri HB, Yasinzadeh M, Kasnavieh SM, Modirian E. Ketamine and atropine for pediatric sedation: a prospective double-blind randomized controlled trial. Pediatr Emerg Care. 2013;29:136-9.
  • 95
    Bar-Joseph G, Guilburd Y, Tamir A, Guilburd JN. Effectiveness of ketamine in decreasing intracranial pressure in children with intracranial hypertension. J Neurosurg Pediatr. 2009;4:40-6.
  • 96
    Zeiler FA, Teitelbaum J, West M, Gillman LM. The ketamine effect on ICP in traumatic brain injury. Neurocrit Care. 2014;21:163-73.
  • 97
    Wang X, Ding X, Tong Y, Zong J, Zhao X, Ren H, et al. Ketamine does not increase intracranial pressure compared with opioids: meta-analysis of randomized controlled trials. J Anesth. 2014;28:821-7.
  • 98
    Zeiler FA, Teitelbaum J, West M, Gillman LM. The ketamine effect on intracranial pressure in nontraumatic neurological illness. J Crit Care. 2014;29:1096-106.
  • 99
    Cohen L, Athaide V, Wickham ME, Doyle-Waters MM, Rose NG, Hohl CM. The effect of ketamine on intracranial and cerebral perfusion pressure and health outcomes: a systematic review. Ann Emerg Med. 2015;65:43-51.
  • 100
    Green SM, Andolfatto G, Krauss BS. Ketamine and intracranial pressure: no contraindication except hydrocephalus. Ann Emerg Med. 2015;65:52-4.
  • 101
    Morgan DJ, Campbell GA, Crankshaw DP. Pharmacokinetics of propofol when given by intravenous infusion. Br J Clin Pharmacol. 1990;30:144-8.
  • 102
    Marik PE. Propofol: therapeutic indications and side-effects. Curr Pharm Des. 2004;10:3639-49.
  • 103
    Godambe SA, Elliot V, Matheny D, Pershad J. Comparison of propofol/fentanyl versus ketamine/midazolam for brief orthopedic procedural sedation in a pediatric emergency department. Pediatrics. 2003;112:116-23.
  • 104
    Mallory MD, Baxter AL, Yanosky DJ, Cravero JP. Pediatric Sedation Research Consortium. Emergency physician-administered propofol sedation: a report on 25,433 sedations from the pediatric sedation research consortium. Ann Emerg Med. 2011;57:462-8.
  • 105
    Srinivasan M, Turmelle M, Depalma LM, Mao J, Carlson DW. Procedural sedation for diagnostic imaging in children by pediatric hospitalists using propofol: analysis of the nature, frequency, and predictors of adverse events and interventions. J Pediatr. 2012;160:801-6.
  • 106
    Sebe A, Yilmaz HL, Koseoglu Z, Ay MO, Gulen M. Comparison of midazolam and propofol for sedation in pediatric diagnostic imaging studies. Postgrad Med. 2014;126:225-30.
  • 107
    Mallory MD, Baxter AL, Kost SI. Pediatric Sedation Research Consortium. Propofol vs pentobarbital for sedation of children undergoing magnetic resonance imaging: results from the Pediatric Sedation Research Consortium. Paediatr Anaesth. 2009;19:601-11.
  • 108
    Pershad J, Wan J, Anghelescu DL. Comparison of propofol with pentobarbital/midazolam/fentanyl sedation for magnetic resonance imaging of the brain in children. Pediatrics. 2007;120:e629-36.
  • 109
    Fang H, Yang L, Wang X, Zhu H. Clinical efficacy of dexmedetomidine versus propofol in children undergoing magnetic resonance imaging: a meta-analysis. Int J Clin Exp Med. 2015;8:11881-9.
  • 110
    Griffiths MA, Kamat PP, McCracken CE, Simon HK. Is procedural sedation with propofol acceptable for complex imaging? A comparison of short vs prolonged sedations in children. Pediatr Radiol. 2013;43:1273-8.
  • 111
    Jalota L, Kalira V, George E, Shi YY, Hornuss C, Radke O, et al. Prevention of pain on injection of propofol: systematic review and meta-analysis. BMJ. 2011;342:d1110.
  • 112
    Chidambaran V, Costandi A, D'Mello A. Propofol: a review of its role in pediatric anesthesia and sedation. CNS Drugs. 2015;29:543-63.
  • 113
    Robinson BJ, Ebert TJ, O'Brien TJ, Colinco MD, Muzi M. Mechanisms whereby propofol mediates peripheral vasodilation in humans. Sympathoinhibition or direct vascular relaxation?. Anesthesiology. 1997;86:64-72.
  • 114
    Jager MD, Aldag JC, Deshpande GG. A presedation fluid bolus does not decrease the incidence of propofol-induced hypotension in pediatric patients. Hosp Pediatr. 2015;5:85-91.
  • 115
    Marik PE. Propofol: therapeutic indications and side effects. Curr Pharm Des. 2004;10:3639-49.
  • 116
    Dosani M, McCormack J, Reimer E, Brant R, Dumont G, Lim J, et al. Slower administration of propofol preserves adequate respiration in children. Paediatr Anaesth. 2010;20:1001-8.
  • 117
    Milius EM, Papademetrious TR, Heitlinger LA. Retrospective review of propofol dosing for procedural sedation in pediatric patients. J Pediatr Pharmacol Ther. 2012;17:246-51.
  • 118
    Murphy A, Campbell DE, Baines D, Mehr S. Allergic reactions to propofol in egg-allergic children. Anesth Analg. 2011;113:140-4.
  • 119
    Chiaretti A, Benini F, Pierri F, Vecchiato K, Ronfani L, Agosto C, et al. Safety and efficacy of propofol administered by paediatricians during procedural sedation in children. Acta Paediatr. 2014;103:182-7.
  • 120
    Vasile B, Rasulo F, Candiani A, Latronico N. The pathophysiology of propofol infusion syndrome: a simple name for a complex syndrome. Intensive Care Med. 2003;29:1417-25.
  • 121
    Alletag MJ, Auerbach MA, Baum CR. Ketamine, propofol, and ketofol use for pediatric sedation. Pediatr Emerg Care. 2012;28:1391-5; quiz 1396-8.
  • 122
    Andolfatto G, Willman E. A prospective case series of pediatric procedural sedation and analgesia in the emergency department using single-syringe ketamine-propofol combination (ketofol). Acad Emerg Med. 2010;17:194-201.
  • 123
    David H, Shipp J. A randomized controlled trial of ketamine/propofol versus propofol alone for emergency department procedural sedation. Ann Emerg Med. 2011;57:435-41.
  • 124
    Yan JW, McLeod SL, Iansavitchene A. Ketamine-propofol versus propofol alone for procedural sedation in the emergency department: a systematic review and meta-analysis. Acad Emerg Med. 2015;22:1003-13.
  • 125
    Jalili M, Bahreini M, Doosti-Irani A, Masoomi R, Arbab M, Mirfazaelian H. Ketamine-propofol combination (ketofol) vs propofol for procedural sedation and analgesia: systematic review and meta-analysis. Am J Emerg Med. 2016;34:558-69.
  • 126
    Miner JR, Moore JC, Austad EJ, Plummer D, Hubbard L, Gray RO. Randomized, double-blinded, clinical trial of propofol, 1:1 propofol/ketamine, and 4:1 propofol/ketamine for deep procedural sedation in the emergency department. Ann Emerg Med. 2015;65:479-88.
  • 127
    Green SM, Andolfatto G, Krauss BS. Ketofol for procedural sedation revisited: pro and con. Ann Emerg Med. 2015;65:489-91.
  • 128
    Belleville JP, Ward DS, Bloor BC, Maze M. Effects of intravenous dexmedetomidine in humans. I. Sedation, ventilation, and metabolic rate. Anesthesiology. 1992;77:1125-33.
  • 129
    Tobias JD. Dexmedetomidine: applications in pediatric critical care and pediatric anesthesiology. Pediatr Crit Care Med. 2007;8:115-31.
  • 130
    Mahmoud M, Mason KP. Dexmedetomidine: review, update, and future considerations of paediatric perioperative and periprocedural applications and limitations. Br J Anaesth. 2015;:115171-82.
  • 131
    Mason KP, Lerman J. Review article: dexmedetomidine in children: current knowledge and future applications. Anesth Analg. 2011;113:1129-42.
  • 132
    Mason KP, Lubisch NB, Robinson F, Roskos R. Intramuscular dexmedetomidine sedation for pediatric MRI and CT. AJR Am J Roentgenol. 2011;197:720-5.
  • 133
    Mekitarian Filho E, Robinson F, de Carvalho WB, Gilio AE, Mason KP. Intranasal dexmedetomidine for sedation for pediatric computed tomography imaging. J Pediatr. 2015;166:1313-5.
  • 134
    Mason KP, O'Mahony E, Zurakowski D, Libenson MH. Effects of dexmedetomidine sedation on the EEG in children. Paediatr Anaesth. 2009;19:1175-83.
  • 135
    Baier NM, Mendez SS, Kimm D, Velazquez AE, Schroeder AR. Intranasal dexmedetomidine: an effective sedative agent for electroencephalogram and auditory brain response testing. Paediatr Anaesth. 2016;26:280-5.
  • 136
    Zhang F, Sun HR, Zheng ZB, Liao R, Liu J. Dexmedetomidine versus midazolam for sedation during endoscopy: a meta-analysis. Exp Ther Med. 2016;11:2519-24.
  • 137
    Carney L, Kendrick J, Carr R. Safety and effectiveness of dexmedetomidine in the pediatric intensive care unit (SAD-PICU). Can J Hosp Pharm. 2013;66:21-7.
  • 138
    Department of Health and Human Services. Public Health Service. Food and Drug Administration. Center for Drug Evaluation and Research. Office of Surveillance and Epidemiology. Pediatric postmarketing pharmacovigilance and drug utilization review. Precedex (dexmedetomidine HCl injection); 2016 Mar. Available from: https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/PediatricAdvisoryCommittee/UCM494470.pdf [cited 18.11.16].
    » https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/PediatricAdvisoryCommittee/UCM494470.pdf
  • 139
    Constantin JM, Momon A, Mantz J, Payen JF, De Jonghe B, Perbet S, et al. Efficacy and safety of sedation with dexmedetomidine in critical care patients: a meta-analysis of randomized controlled trials. Anaesth Crit Care Pain Med. 2016;35:7-15.
  • 140
    Berkenbosch JW, Wankum PC, Tobias JD. Prospective evaluation of dexmedetomidine for noninvasive procedural sedation in children. Pediatr Crit Care Med. 2005;6:435-9, quiz 440.
  • 141
    Forman SA. Clinical and molecular pharmacology of etomidate. Anesthesiology. 2011;114:695-707.
  • 142
    Falk J, Zed PJ. Etomidate for procedural sedation in the emergency department. Ann Pharmacother. 2004;38:1272-7.
  • 143
    Mandt MJ, Roback MG, Bajaj L, Galinkin JL, Gao D, Wathen JE. Etomidate for short pediatric procedures in the emergency department. Pediatr Emerg Care. 2012;28:898-904.
  • 144
    Di Liddo L, D'Angelo A, Nguyen B, Bailey B, Amre D, Stanciu C. Etomidate versus midazolam for procedural sedation in pediatric outpatients: a randomized controlled trial. Ann Emerg Med. 2006;48:433-40, 440.e1.
  • 145
    Majesko A, Darby JM. Etomidate and adrenal insufficiency: the controversy continues. Crit Care. 2010;14:338.
  • 146
    Wagner RL, White PF, Kan PB, Rosenthal MH, Feldman D. Inhibition of adrenal steroidogenesis by the anesthetic etomidate. N Engl J Med. 1984;310:1415-21.
  • 147
    Jones AE. The etomidate debate. Ann Emerg Med. 2010;56:490-1.
  • 148
    Tekwani KL, Watts HF, Sweis RT, Rzechula KH, Kulstad EB. A comparison of the effects of etomidate and midazolam on hospital length of stay in patients with suspected sepsis: a prospective, randomized study. Ann Emerg Med. 2010;56:481-9.

Publication Dates

  • Publication in this collection
    2017

History

  • Received
    4 May 2017
  • Accepted
    26 May 2017
Sociedade Brasileira de Pediatria Av. Carlos Gomes, 328 cj. 304, 90480-000 Porto Alegre RS Brazil, Tel.: +55 51 3328-9520 - Porto Alegre - RS - Brazil
E-mail: jped@jped.com.br