Over the past 10 years, there has been an increase in

Similar documents
Use of Dexmedetomidine for Sedation of Children Hospitalized in the Intensive Care Unit

Susan Becker DNP, RN, CNS, CCRN, CCNS Marymount University, Arlington, VA

ASMIC 2016 DEXMEDETOMIDINE IN THE INTENSIVE CARE UNIT DR KHOO TIEN MENG

DOI /yydb medetomidine a review of clinical applications J. Curr Opin Anaesthesiol

Dexmedetomidine. Dr.G.K.Kumar,M.D.,D.A., Assistant Professor, Madras medical college,chennai. History

Therapeutics and clinical risk management (2011) Vol.7:291~299. Dexmedetomidine hydrochloride as a long-term sedative.

Comparison of dexmedetomidine and propofol in mechanically ventilated patients with sepsis: A pilot study

Disclosures. Dexmedetomidine: The Good, The Bad and The Delirious. The Delirious. Objectives. Characteristics of Delirium. Definition of Delirium

Appendix: Outcomes when Using Adjunct Dexmedetomidine with Propofol Sedation in

A COMPARATIVE STUDY OF MIDAZOLAM, PROPOFOL AND DEXMEDETOMIDINE INFUSIONS FOR SEDATION IN ME- CHANICALLY VENTILATED PATIENTS IN ICU

Propofol vs Dexmedetomidine

Dexmedetomidine use in a pediatric cardiac intensive care unit: Can we use it in infants after cardiac surgery?

Invasive and noninvasive procedures

Corresponding author: V. Dua, Department of Anaesthesia, BJ Wadia Hospital for Children, Parel, Mumbai, India.

Clinical effectiveness of a sedation protocol minimizing benzodiazepine infusions and favoring early dexmedetomidine: A before-after study

Dexmedetomidine and its Injectable Anesthetic-Pain Management Combinations

Dexmedetomidine for Sedation in the Critical Care Setting: An Economic Assessment

NIH Public Access Author Manuscript J Crit Care. Author manuscript; available in PMC 2013 July 28.

Original Contributions

Dexmedetomidine Versus Midazolam for the Sedation of Patients with Non-invasive Ventilation Failure

The Addition of Dexmedetomidine as an Adjunctive Therapy to Benzodiazepine Use in Alcohol Withdrawal Syndrome

PDF of Trial CTRI Website URL -

Alfaxan. (alfaxalone 10 mg/ml) Intravenous injectable anesthetic for use in cats and dogs. TECHNICAL NOTES DESCRIPTION INDICATIONS

TITLE: Dexmedetomidine for Sedation in the ICU or PICU: A Review of Cost- Effectiveness and Guidelines

What dose of methadone should I use?

Evaluation of dexmedetomine in anesthesia care for elderly patients with obstructive sleep apnea

TITLE: Dexmedetomidine for Sedation of Patients in the ICU or PICU: Review of Clinical Effectiveness and Safety

Preliminary UK experience of dexmedetomidine, a novel agent for postoperative sedation in the intensive care unit

PAEDIATRIC DEXMEDETOMIDINE INFUSIONS IN BURNS INTENSIVE CARE

Dexmedetomidine for Sedation of Neonates with HIE Undergoing Therapeutic Hypothermia: A Single-Center Experience

The evolving approach to sedation in ventilated patients: a real world perspective

Neonates and infants undergoing radiological imaging

Original Article Dose-dependent effects of dexmedetomidine during one-lung ventilation in patients undergoing lobectomy

A New Advancement in Anesthesia. Your clear choice for induction.

Period of study: 12 Nov 2002 to 08 Apr 2004 (first subject s first visit to last subject s last visit)

Top 5 Short Procedure Sedation Scenarios

Original Article Different doses of dexmedetomidine in children with non-tracheal intubation intravenous general anesthesia

Hemodynamic effects of dexmedetomidine-- fentanyl vs. nalbuphine--propofol in plastic surgery

ISMP Canada HYDROmorphone Knowledge Assessment Survey

Dexmedetomidine, an 2 adrenergic agonist, was

Int. J. Pharm. Sci. Rev. Res., 36(1), January February 2016; Article No. 38, Pages: *Corresponding author s

T u l a n e U n i v e r s i t y I A C U C Guidelines for Rodent & Rabbit Anesthesia, Analgesia and Tranquilization & Euthanasia Methods

Clinical applicability of dexmedetomidine for sedation, premedication and analgesia in cats 1 / 2007

Perioperative Care of Swine

Comparison of Intensive Care Unit Sedation Using Dexmedetomidine, Propofol, and Midazolam

Department of clinical pathology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt

GUIDELINES FOR ANESTHESIA AND FORMULARIES

SCIENTIFIC COOPERATIONS MEDICAL WORKSHOPS July, 2015, Istanbul - TURKEY

Case Report Dexmedetomidine as a Procedural Sedative for Percutaneous Tracheotomy: Case Report and Systematic Literature Review

Dexmedetomidine intravenous sedation using a patient-controlled sedation infusion pump: a case report

Review Article Clinical efficacy of dexmedetomidine versus propofol in children undergoing magnetic resonance imaging: a meta-analysis

Quality of MRI pediatric sedation: Comparison between intramuscular and intravenous dexmedetomidine

A SYSTEMATIC REVIEW ON THE USE OF DEXMEDETOMIDINE AS A SOLE AGENT FOR INTRAVENOUS MODERATE SEDATION

Original Article Effects of low dose midazolam on bradycardia and sedation during dexmedetomidine infusion

Dexmedetomidine: its use in intensive care medicine and anaesthesia

Ashraf Darwish, Rehab Sami, Mona Raafat, Rashad Aref and Mohamed Hisham

Anesthesia in an English Bulldog

Dexmedetomidine vs. Propofol for Short-Term Sedation of Postoperative Mechanically Ventilated Patients

Premedication with alpha-2 agonists procedures for monitoring anaesthetic

Comparison of two doses of intranasal dexmedetomidine as premedication in children

Pain Management in Racing Greyhounds

A Clinical Study of Dexmedetomidine under Combined Spinal Epidural Anaesthesia at a Tertiary Care Hospital

Diskography is a diagnostic modality used to

The Aquila Digital Community. The University of Southern Mississippi. Benjamin Heinrich Riebesel University of Southern Mississippi

Does Early and Appropriate Antibiotic Administration Improve Mortality in Emergency Department Patients with Severe Sepsis or Septic Shock?

DOES TIMING OF ANTIBIOTICS IMPACT OUTCOME IN SEPSIS? Saravana Kumar MD HEAD,DEPT OF EM,DR MEHTA S HOSPITALS CHENNAI,INDIA

Day 90 Labelling, PL LABELLING AND PACKAGE LEAFLET

August 16, Implementing High Quality, High Volume Spay/Neuter: Challenges & Solutions

Dexmedetomidine for Emergence Agitation after Sevoflurane Anesthesia in Preschool Children Undergoing Day Case Surgery: Comparative Dose-Ranging Study

Sedative and antinociceptive effects of dexmedetomidine and buprenorphine after oral transmucosal or intramuscular administration in cats

Procedure # IBT IACUC Approval: December 11, 2017

Associate Professor, Department of Anaesthesiology, Government Thoothukudi Medical College, Thoothukudi, Tamil Nadu, India, 2

Anesthetic regimens for mice, rats and guinea pigs

Evaluation of efficacy of sedative and analgesic effects of single IV dose of dexmedetomidine in post-operative patients

N.C. A and T List of Approved Analgesics 1 of 5

Dıfferent Doses Of Dexmedetomidine On Controllıng Haemodynamıc Responses To Tracheal Intubatıon

Associate Professor, Department of Anaesthesiology, Rangaraya Medical College, Kakinada, East Godavari, Andhra Pradesh, India, 2

Inappropriate Use of Antibiotics and Clostridium difficile Infection. Jocelyn Srigley, MD, FRCPC November 1, 2012

Study the Effect of Dexmedetomidine on Emergence Agitation after Nasal Surgeries

Pharmacoeconomic analysis of selected antibiotics in lower respiratory tract infection Quenzer R W, Pettit K G, Arnold R J, Kaniecki D J

Sedation and analgesia in patients on mechanical ventilation in pediatric intensive care units in Argentina

Dexmedetomidine: review, update, and future considerations of paediatric perioperative and periprocedural applications and limitations

DISSOCIATIVE ANESTHESIA

A Comparative Evaluation of Intranasal Dexmedetomidine and Intranasal Midazolam for Premedication in Pediatric Surgery

Rajaclimax Kirubahar, Bose Sundari, Vijay Kanna*, Kanakasabai Murugadoss

Alfaxalone induction dose following administration of medetomidine and butorphanol in the dog

Role of Dexmedetomidine as an Anesthetic Adjuvant in Laparoscopic Surgery

Welcome! 10/26/2015 1

SUMMARY OF PRODUCT CHARACTERISTICS

CLINICAL ESSENTIAL HUDDLE CARD. All associates must comply with their state practice acts.

Comparison of dexmedetomidine and propofol for conscious sedation in inguinal hernia repair: A prospective, randomized, controlled trial

2018/19 VETERINARY TECHNICIAN CERTIFICATE COURSE IN PRINCIPLE TECHNIQUES OF SMALL ANIMAL ANESTHESIA, PERIOPERATIVE ANALGESIA & CRITICAL PATIENT CARE

Safety and Efficacy of Dexmedetomidine, Ketofol, and Propofol for Sedation of Mechanically Ventilated Patients

APPLICATION FOR LIVE ANIMAL USE IN TEACHING AT FAULKNER STATE COMMUNITY COLLEGE

Evaluating the Role of MRSA Nasal Swabs

Methicillin-Resistant Staphylococcus aureus Nasal Swabs as a Tool in Antimicrobial Stewardship

NUMBER: R&C-ARF-10.0

Alfaxan FAQs. Repeatable. Reliable. Relax.

Module C Veterinary Anaesthesia Small Animal Anaesthesia and Analgesia (C-VA.1)

Comparison of dexmedetomidine v/s propofol used as adjuvant with combined spinal epidural anaesthesia for joint replacement surgeries

Transcription:

Dexmedetomidine for Sedation During Noninvasive Ventilation in Pediatric Patients Rasika Venkatraman, MD 1 ; James L. Hungerford, MD 2,3 ; Mark W. Hall, MD 1 ; Melissa Moore-Clingenpeel, MS 1,4 ; Joseph D. Tobias, MD 1,5,6 1 Division of Critical Care, Department of Pediatrics, Nationwide Children s Hospital and The Ohio State University College of Medicine, Columbus, OH. 2 Emory Sleep Center, Emory University, Atlanta, GA. 3 Critical Care Medicine, Children's Healthcare of Atlanta, Children s at Egleston, Atlanta, GA. 4 Biostatistics Core, The Research Institute at Nationwide Children s Hospital, Columbus, OH. 5 Department of Anesthesiology & Pain Medicine, Nationwide Children s Hospital, Columbus, OH. 6 Department of Anesthesiology & Pain Medicine, The Ohio State University College of Medicine, Columbus, OH. Drs. Venkatraman, Hall, and Moore-Clingenpeel disclosed off-label product use of dexmedetomidine for longer durations than are currently approved by the Food and Drug Administration. The remaining authors have disclosed that they do not have any potential conflicts of interest. For information regarding this article, E-mail: Joseph.Tobias@Nationwide- Childrens.org Copyright 2017 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies DOI: 10.1097/PCC.0000000000001226 Objectives: To describe the use of dexmedetomidine for sedation in a large cohort of nonintubated children with acute respiratory insufficiency receiving noninvasive ventilatory support. Design: Single-center, retrospective, observational cohort study. Setting: A large quaternary-care PICU. Patients: The study cohort included 202 children receiving noninvasive ventilatory and a dexmedetomidine infusion within 48 hours of PICU admission over a 6-month period. Interventions: None. Measurements and Main Results: The primary respiratory diagnoses in the cohort (median age, 2 yr) included status asthmaticus (60%) and bronchiolitis (29%). Dexmedetomidine was infused for a median of 35 hours with a median hourly dose across the patient cohort of 0.61 μg/kg/hr (range, 0.4 0.8 μg/kg/hr). The target sedation level was achieved in 168 patients (83%) in the cohort for greater than or equal to 80% of the recorded values over the entire noninvasive ventilatory course, with dexmedetomidine as the only continuously administered sedative agent. While vital signs were frequently abnormal relative to age-based norms, clinical interventions were needed rarely to treat bradycardia (13%), hypotension (20%), and hypopnea (5%). The most frequently used of these interventions was a dexmedetomidine dose reduction, fluid bolus, and titration of noninvasive ventilatory support. Five patients (2.5%) required endotracheal intubation: three due to progression of their respiratory illness, one with septic shock, and one with apnea requiring resuscitation. In 194 of 202 patients (96%), the outcome of the noninvasive ventilatory course was successful with the patient being weaned from noninvasive respiratory support to nasal cannula or room air. Conclusions: Dexmedetomidine was often effective as a single continuous sedative infusion during pediatric noninvasive ventilatory. Cardiorespiratory events associated with its use were typically mild and/or reversible with dose reduction, fluid administration, and/or noninvasive ventilatory titration. Prospective studies comparing dexmedetomidine with other agents in this setting are warranted. (Pediatr Crit Care Med 2017; XX:00 00) Key Words: dexmedetomidine; noninvasive ventilation; sedation Over the past 10 years, there has been an increase in the use of noninvasive ventilation (NIV) including biphasic positive pressure ventilation (BiPAP) and continuous positive airway pressure (CPAP) ventilation in children with respiratory insufficiency. This therapy allows for the provision of respiratory support while avoiding endotracheal intubation in many patients. The use of this technique requires the application of a tight-fitting mask and necessitates synchronous breathing with the device. Many pediatric patients, particularly young children, require sedation in order to effectively tolerate this process. Dexmedetomidine (Precedex, Hospira Worldwide, Lake Forest, IL) is frequently used in our PICU as the first-line agent for sedation in this patient population. Dexmedetomidine is the pharmacologically active dextro-isomer of medetomidine, exerting its physiologic effects via α 2 -adrenergic receptors (1). Its effects include anxiolysis and sedation with minimal effects on respiratory function and upper airway dynamics (1 6). Although there are published data in the adult population, there are no reports regarding the use of dexmedetomidine for sedation during NIV in pediatric patients (7 9). We therefore designed a single-center, retrospective study to test the hypotheses that 1) dexmedetomidine as a single continuous agent would be sufficient to achieve the target sedation level in the Pediatric Critical Care Medicine www.pccmjournal.org 1

Venkatraman et al majority of children receiving NIV, 2) adverse cardiorespiratory events including bradycardia, hypotension, and respiratory depression will occur infrequently, and 3) these adverse events would have limited clinical implications and be reversible with dose reduction. METHODS This retrospective chart review was approved by the Institutional Review Board of Nationwide Children s Hospital, Columbus, Ohio (IRB13-00334), and was registered at ClinicalTrials.gov. Data were reviewed in the electronic medical record by a single reviewer (R.V.) and were cross-checked by other authors for reliability. All admissions to our 40-bed quaternary care PICU over a 6-month period were screened. Inclusion criteria were PICU admission for respiratory insufficiency and initiation of both NIV and a dexmedetomidine infusion within 48 hours of PICU admission. The definition of NIV included the use of CPAP, BiPAP, or high-flow nasal cannula. Exclusion criteria included age greater than or equal to 18 years, endotracheally intubated patients, patients with a tracheostomy, and patients who were on NIV and dexmedetomidine for less than 3 hours. Demographic data collected included age, gender, primary diagnosis causing respiratory insufficiency, hospital length of stay (LOS), and PICU LOS. To test the first part of our hypothesis relating to achievement of target sedation score, we collected the following information: starting dose of dexmedetomidine, any dose adjustments, reason for dose adjustment (if known), and the total duration of infusion (hr). We also documented the use of other continuous or intermittent sedative drugs. Data were collected for each patient only for the first 7 days after starting dexmedetomidine. If the duration of the infusion was shorter than 7 days, data were collected until either the dexmedetomidine infusion and/or NIV was discontinued or the patient required endotracheal intubation for respiratory failure. The adequacy of sedation was assessed using the State Behavioral Scale (SBS) (10) (Fig. 1) which is routinely measured in our PICU every 4 hours during continuous sedation, with a typical target SBS score of 0 to 1 while on NIV. To test the second and third parts of our hypothesis related to adverse cardiorespiratory events associated with dexmedetomidine use, we collected physiologic data including heart rate, blood pressure, and respiratory rate. Bradycardia, hypotension, and hypopnea were defined as values less than the 5th percentile for age as defined by Pediatric Acute Life Support (PALS) criteria (www.cpr.heart.org). The extremes of vital signs were recorded in 4-hour epochs while on dexmedetomidine. Data were also collected on interventions that were performed to manage these events. Clinically significant events in the case of bradycardia and hypotension were defined as those treated with discontinuation or dose reduction of dexmedetomidine, fluid boluses, vasopressors, or cardiopulmonary resuscitation (CPR). Additionally, hypopnea associated with O 2 desaturation less than 90% and/or a need for titration of NIV was considered clinically significant. Data are expressed as the median with interquartile ranges, as data were nonparametrically distributed. Proportions were expressed in percentages. Data were analyzed using the SAS statistical software (SAS institute, Cary, NC). RESULTS Three hundred ninety-six patients received a dexmedetomidine infusion upon admission to the PICU during the study period. Of these, 202 patients met inclusion criteria and were included in the analysis (Fig. 2). Ninety-seven of the 202 patients (48%) were male and 105 (52%) were female. Primary diagnoses necessitating NIV therapy included status asthmaticus (60%), bronchiolitis (29%), or septic shock and other (11%). The demographic characteristics are summarized in Table 1. In 194 of 202 patients (96%), the outcome of the NIV course was weaning from noninvasive respiratory support to nasal cannula or room air within 7 days. Five of 202 patients (2.5%) required endotracheal intubation. Three patients (1.5%) remained on NIV at the end of the 7-day period of data collection. Dexmedetomidine was infused for a median of 35 hours (17.5 51.5 hr). The minimum duration of the infusion was 4.5 hours and the maximum was 652 hours. The median average hourly dose across the patient cohort was 0.61 μg/kg/hr (0.4 0.8 μg/kg/hr) with a maximum infusion rate of 1.5 μg/kg/hr. One hundred forty of the 202 patients (69%) received dexmedetomidine for more than 24 hours and 74 patients (37% of the total cohort of 202) received dexmedetomidine for more than 48 hours. Documentation of measured and target sedation scores was present for 2,039 of 2,872 expected observation points (71%) among the cohort as a whole. Eleven patients (5.4%) had no target or actual SBS scores documented. One hundred sixtyeight patients (83% of the full cohort) achieved the target sedation level (SBS, 0 to 1) for at least 80% of recorded values over the entire NIV course. Of these patients, 146 achieved the target sedation level at least 80% of the time with the dexmedetomidine infusion alone. The remaining patients who successfully achieved target sedation level needed at least one dose of an additional agent for sedation, but most were intermittent doses. A minority of patients (7% of the cohort) received additional sedative infusions. Among the 158 patients for whom documentation of measured and target SBS scores throughout the first 8 hours of dexmedetomidine infusion were available, 101 (64%) achieved the target level of sedation level (SBS, 0 to 1) with dexmedetomidine as a single continuous agent. An additional 24 of the 158 patients were able to achieve the target sedation level within 8 hours with dexmedetomidine and at least one dose of an intermittent sedative agent. Of the 106 patients (52% of the entire cohort) who received at least one dose of an adjunctive sedative medication at some point during the NIV course, the majority required only one or two doses, whereas 65 patients (32%) required greater than or equal to five doses of additional intermittent sedative agents. Of those patients who required greater than or equal to five doses of an additional agent, 45 (69%) were on the dexmedetomidine infusion for greater than or equal to 48 hours. The most 2 www.pccmjournal.org XXX 2017 Volume XX Number XXX

Feature Article Figure 1. State Behavioral Scale (SBS) (10). ETT = endotracheal tube. Pediatric Critical Care Medicine www.pccmjournal.org 3

Venkatraman et al TABLE 1. Demographic Information of the Study Cohort Demographic Feature Demographic Data Figure 2. Screening and enrollment summary for the study cohort. DEX = dexmedetomidine, NIV = noninvasive ventilatory. common supplemental sedative agent used was IV lorazepam (59%). Other agents used as adjuncts to the dexmedetomidine infusion included IV midazolam (44%) and enteral chloral hydrate (39%). Fourteen subjects (7%) received continuous IV infusions of other sedative/analgesic drugs including IV midazolam (n = 6, 3%) and hydromorphone (n = 5, 2.5%) in addition to dexmedetomidine. When evaluating vital sign changes relative to age-based norms, bradycardia occurred in 26 patients (13%), hypotension in 56 patients (28%), and hypopnea in 136 patients (67%) at some time during the dexmedetomidine course. After applying the a priori definitions for clinically relevant events, clinically significant bradycardia was seen in 26 patients (13%), clinically significant hypotension in 41 patients (20%), and clinically significant hypopnea in only 11 patients (5%) (Table 2). Among those who received an intervention for an adverse event, the most common was dose reduction or discontinuation of the dexmedetomidine infusion. All 26 patients who met criteria for bradycardia for age received a dexmedetomidine dose reduction or discontinuation as their only intervention. Of the 41 patients who had clinically relevant hypotension, fluid boluses and/or dexmedetomidine dose adjustment were common. The vast majority of hypopnea Age, 2.0 (1.0 4.0) < 6 mo, n (%) 16 (7.9) 6 mo to 1 yr, n (%) 30 (14.9) 1 2 yr, n (%) 38 (18.8) > 2 yr, n (%) 118 (58.4) Primary diagnosis, n (%) Status asthmaticus 122 (60.4) Acute bronchiolitis 59 (29.2) Malignancy or other oncologic diagnosis 11 (5.4) Croup 5 (2.5) Septic shock 5 (2.5) Admit weight (kg), 13.6 (9.8 20.8) PICU length of stay (d), Hospital length of stay (d), Cumulative total dose of dexmedetomidine, μg/kg, Total hours on dexmedetomidine, Total days on dexmedetomidine, IQR = interquartile range. 3 (2 5) 4 (3 6) 20.43 (9.2 32.2) 35.0 (17.5 51.5) 1.5 (0.7 2.2) events did not require clinical intervention. Eleven patients exhibited transient desaturation in the setting of hypopnea that was managed with titration of NIV support. One patient, a 1-month-old infant with bronchiolitis, experienced apnea with progression to bradycardic arrest. This was the only patient in the cohort (1/202 or 0.5%) who received CPR and vasopressor therapy. This patient received fluid boluses, endotracheal intubation, and the dexmedetomidine infusion was discontinued. Four additional patients (2%) underwent endotracheal intubation: three with progression of their respiratory illness (tachypnea and respiratory distress) and one with septic shock (tachycardia with poor perfusion). There were no deaths among the patients in the study. DISCUSSION Dexmedetomidine is commonly used in our combined medical/surgical PICU as a continuous infusion to provide sedation and anxiolysis in critically ill, spontaneously breathing infants and children with respiratory insufficiency to facilitate tolerance of NIV. This represents the largest report to date of 4 www.pccmjournal.org XXX 2017 Volume XX Number XXX

Feature Article TABLE 2. Adverse Cardiorespiratory Events During Dexmedetomidine Infusion Adverse Event Bradycardia Hypotension Hypopnea Cardiorespiratory Arrest Overall frequency 26/202 (13%) 56/202 (28%) 136 (67%) 1/202 (0.5%) Event required intervention 26/202 (13%) 41/202 (20%) 11/202 (5%) 1/202 (0.5%) Intervention Dexmedetomidine dose reduction 26/202 (13%) 25/202 (12%) a 0/202 (0%) 1/202 (0.5%) Fluid bolus 0/202 (0%) 32/202 (16%) a 0/202 (0%) 1/202 (0.5%) Vasoactive drug 0/202 (0%) 0/202 (0%) 0/202 (0%) 1/202 (0.5%) Titration of respiratory support 0/202 (0%) 0/202 (0%) 11/202 (5%) 1/202 (0.5%) Cardiopulmonary resuscitation 0/202 (0%) 0/202 (0%) 0/202 (0%) 1/202 (0.5%) a Sixteen subjects received both dose reduction and fluid bolus. dexmedetomidine use in nonintubated children during noninvasive positive pressure ventilation. Dexmedetomidine is a selective α 2 -adrenergic receptor agonist that possesses analgesic and sedative properties, with minimal tendency for respiratory depression. Current uses described in the literature include sedation in the intensive care setting, and as an adjunctive agent for procedural sedation and analgesia (1). Its use in the setting of postoperative cardiac surgery has been well described in children, including infants and neonates (11 14). The use of dexmedetomidine was recently reported in a cohort of more than 1,000 intubated children with acute respiratory failure with the conclusion that it works well as a primary sedative agent in those with low criticality and may facilitate tracheal extubation (15). The therapeutic effects of dexmedetomidine are mediated throughout the central nervous system, with the sedative and anxiolytic effects resulting from its activity in the locus ceruleus and the analgesic effects from the dorsal horn of the spinal cord (16). Because of its sympatholytic activity, the most common adverse effects are bradycardia and hypotension. Notable beneficial effects, which may be particularly relevant in the setting of NIV, include its potential to decrease airway reactivity and limit the frequency of delirium in the ICU setting. Groeben et al (17) suggested that dexmedetomidine infusion may have a therapeutic effect in bronchospasm and may decrease airway reactivity, based on the results of their animal study with histamine-induced bronchoconstriction. In addition, dexmedetomidine may be less likely to cause delirium when compared with benzodiazepines such as midazolam and has been shown to not disrupt sleep cycles, facilitating nonrapid eye movement sleep patterns (18, 19). For these reasons, dexmedetomidine is an attractive agent for use in the spontaneously breathing patient in whom control of agitation without respiratory depression remains a key clinical goal. The literature on the use of dexmedetomidine during NIV is relatively sparse and is limited to adult populations. Case series have demonstrated that dexmedetomidine can safely resolve acute agitation during NIV therapy with limited and reversible adverse effects on hemodynamic function (7, 8). A small prospective study of 10 adult patients using dexmedetomidine during NIV reported that all patients achieved target sedation score within 1 hour of initiation of the infusion without substantial hemodynamic changes (9). Similar results were reported in a subsequent randomized trial of 40 adult patients with acute agitation during NIV therapy (20). Dexmedetomidine and midazolam infusions were equivalent in their ability to maintain patients at the desired level of sedation and maintain hemodynamic stability. A more recent randomized trial of 62 adult patients with cardiogenic pulmonary edema found that sedation with dexmedetomidine during NIV was associated with a lower need for endotracheal intubation, shorter ICU LOS, and shorter time of mechanical ventilation (4). Despite its popularity and the large clinical experience in the pediatric population, dexmedetomidine has Food and Drug Administration approval only for use in adults for up to 24 hours either for sedation during mechanical ventilation or as an agent for sedation during monitored anesthesia care. The majority of data concerning the pediatric applications of dexmedetomidine remain centered on its use for short durations (< 72 hr) and in endotracheally intubated, mechanically ventilated patients (1, 2, 5, 6, 11 15). We report the use of dexmedetomidine for sedation during NIV in 202 spontaneously breathing, critically ill infants and children with acute respiratory insufficiency. Using moderate-dose dexmedetomidine infusions (median average hourly dose of 0.61 μg/kg/hr), two thirds of the patients (64%) achieved target sedation level with dexmedetomidine as a single continuous agent alone within 8 hours of initiation of the infusion. Eighty-three percent were able to achieve target sedation level with dexmedetomidine as the primary continually infused sedative agent, allowing for the use of intermittent sedatives, usually lorazepam, during the NIV course. As many patients did not receive a bolus dose of dexmedetomidine, one third of the patients in this series required more than five additional doses of adjunctive agents (lorazepam, midazolam, chloral hydrate, or hydromorphone), especially during the first 4 6 hr of therapy with NIV. Most patients (96%) had a successful outcome of the NIV course and were eventually weaned from NIV to nasal cannula or room air with failure of NIV occurring rarely (5/202, 2.5%). Pediatric Critical Care Medicine www.pccmjournal.org 5

Venkatraman et al Vital sign abnormalities, as defined using PALS criteria, were noted with bradycardia occurring in 13% of subjects, hypotension in 28%, and respiratory depression or hypopnea in 67%. Clinically relevant vital sign changes were rare and typically manageable with dexmedetomidine dose reduction, fluid administration, or titration of NIV. The one patient in the cohort who had apnea and a bradycardic arrest while on dexmedetomidine was a 1-month-old infant with bronchiolitis. Although the apnea episode was felt to be related to the subject s underlying disease process, it is possible that dexmedetomidine may have been contributory. Eventually, the patient underwent successful tracheal extubation and had a favorable outcome. The goals of sedation during NIV vary somewhat from those during endotracheal intubation and mechanical ventilation. During NIV, the goals include preservation of the day-night cycle in the hopes of limiting delirium, tolerance of the external device (mask or nasal prongs), and improvement of synchrony with respiratory support. These must be achieved while avoiding deleterious effects on upper airway patency, diaphragmatic function, and central control of ventilation. Given the paucity of evidence-based medicine, there are limited data in the pediatric population demonstrating the superiority of one agent over another (21). In clinical practice, various agents or combinations of agents have been used to provide sedation during NIV including opioids, benzodiazepines, ketamine, and dexmedetomidine. Concerns with an increased frequency of delirium may limit the use of benzodiazepines in this and other clinical scenarios (18, 19). As opioids and benzodiazepines may impact upper airway patency and central control of ventilation, dexmedetomidine and ketamine may be the preferred options (22, 23). Although ketamine has a limited impact on respiratory function at doses used for procedural sedation and may improve respiratory dynamics (decreased airway resistance, improved dynamic compliance, preservation of functional residual capacity), ketamine may increase airway secretions and lead to delirium and emergence reactions (23, 24). The retrospective nature of the study represents a major limitation. A retrospective analysis likely results in the significant underreporting of physiologic changes, clinical events, and clinician responses. As such, the information provided can only be accepted as preliminary data which must be further studied in prospective trials with head-to-head comparisons with other agents. It is impossible to differentiate the effects of dexmedetomidine from other causes of bradycardia, hypotension, or hypopnea (e.g., the patient s underlying condition, other medications, or adjunct sedative medications). Our results may not be generalizable to all critically ill children given that we did not include neonatal or cardiothoracic ICU patients. Also, documentation of actual and target sedation scores was unavailable for approximately 30% of the 4-hour epochs included in this study. This limitation is mitigated somewhat by the fact that 95% of subjects did have sedation scoring performed, albeit less consistently than intended. Furthermore, as the protocol for the use of dexmedetomidine was not controlled, there was significant variability in the clinical practice involving the decision to initiate and titrate NIV as well as the choice of agent for sedation. Given the limited variability in our clinical practice, dexmedetomidine is the primary agent used for sedation. However, variability existed with the choice of a supplemental (secondary) agent when adjunctive sedation was needed. This is the first large-scale report of dexmedetomidine as the primary sedative agent for the specific indication of tolerance of NIV in spontaneously breathing, critically ill infants and children. The outcomes and findings of our study mirror the findings of both the adult and pediatric literature with respect to the hemodynamic side effect profile of dexmedetomidine. The results are encouraging, with most patients successfully completing their NIV course, and a low rate of clinically significant cardiorespiratory adverse events. Seventy-eight of the patients (39%) in the study manifested changes in vital signs that met our thresholds for identification of potential adverse effects. When required, treatment was generally limited to a decrease in the infusion rate or a fluid bolus. As interventions to treat dexmedetomidine-induced vital sign abnormalities were occasionally required, we believe that this supports the need for ICU-level monitoring in this population. As with other sedative agents, tolerance occurs with dexmedetomidine and strategies must be employed for tapering (weaning) the infusion or a transition to other agents (clonidine) following its use for more than 72 hours (25, 26). With these caveats in mind, we noted that dexmedetomidine was effective as the primary continuously administered sedative agent during NIV. Future prospective studies are needed to provide additional information regarding the efficacy and adverse effect profile of dexmedetomidine compared with other commonly used sedative agents such as midazolam. REFERENCES 1. Tobias JD: Dexmedetomidine: Applications in pediatric critical care and pediatric anesthesiology. Pediatr Crit Care Med 2007; 8:115 131 2. Mahmoud M, Jung D, Salisbury S, et al: Effect of increasing depth of dexmedetomidine and propofol anesthesia on upper airway morphology in children and adolescents with obstructive sleep apnea. J Clin Anesth 2013; 25:529 541 3. Ankichetty S, Wong J, Chung F: A systematic review of the effects of sedatives and anesthetics in patients with obstructive sleep apnea. J Anaesthesiol Clin Pharmacol 2011; 27:447 458 4. Huang Z, Chen YS, Yang ZL, et al: Dexmedetomidine versus midazolam for the sedation of patients with non-invasive ventilation failure. Intern Med 2012; 51:2299 2305 5. Zhuang PJ, Wang X, Zhang XF, et al: Postoperative respiratory and analgesic effects of dexmedetomidine or morphine for adenotonsillectomy in children with obstructive sleep apnoea. Anaesthesia 2011; 66:989 993 6. Mason KP, Lerman J: Review article: Dexmedetomidine in children: Current knowledge and future applications. Anesth Analg 2011; 113:1129 1142 7. Duan M, Lee J, Bittner EA: Dexmedetomidine for sedation in the parturient with respiratory failure requiring noninvasive ventilation. Respir Care 2012; 57:1967 1969 8. Takasaki Y, Kido T, Semba K: Dexmedetomidine facilitates induction of noninvasive positive pressure ventilation for acute respiratory failure in patients with severe asthma. J Anesth 2009; 23:147 150 6 www.pccmjournal.org XXX 2017 Volume XX Number XXX

Feature Article 9. Akada S, Takeda S, Yoshida Y, et al: The efficacy of dexmedetomidine in patients with noninvasive ventilation: A preliminary study. Anesth Analg 2008; 107:167 170 10. Curley MA, Harris SK, Fraser KA, et al: State Behavioral Scale: A sedation assessment instrument for infants and young children supported on mechanical ventilation. Pediatr Crit Care Med 2006; 7:107 114 11. Bejian S, Valasek C, Nigro JJ, et al: Prolonged use of dexmedetomidine in the paediatric cardiothoracic intensive care unit. Cardiol Young 2009; 19:98 104 12. Berkenbosch JW: Dexmedetomidine and pediatric (cardiac) critical care are we there yet? Pediatr Crit Care Med 2010; 11:148 149 13. Chrysostomou C, Di Filippo S, Manrique AM, et al: Use of dexmedetomidine in children after cardiac and thoracic surgery. Pediatr Crit Care Med 2006; 7:126 131 14. Chrysostomou C, Sanchez De Toledo J, Avolio T, et al: Dexmedetomidine use in a pediatric cardiac intensive care unit: Can we use it in infants after cardiac surgery? Pediatr Crit Care Med 2009; 10:654 660 15. Grant MJ, Schneider JB, Asaro LA, et al; Randomized Evaluation of Sedation Titration for Respiratory Failure Study Investigators: Dexmedetomidine use in critically ill children with acute respiratory failure. Pediatr Crit Care Med 2016; 17:1131 1141 16. Guinter JR, Kristeller JL: Prolonged infusions of dexmedetomidine in critically ill patients. Am J Health Syst Pharm 2010; 67: 1246 1253 17. Groeben H, Mitzner W, Brown RH: Effects of the alpha2-adrenoceptor agonist dexmedetomidine on bronchoconstriction in dogs. Anesthesiology 2004; 100:359 363 18. Pandharipande P, Shintani A, Peterson J, et al: Lorazepam is an independent risk factor for transitioning to delirium in intensive care unit patients. Anesthesiology 2006; 104:21 26 19. Riker RR, Shehabi Y, Bokesch PM, et al; SEDCOM (Safety and Efficacy of Dexmedetomidine Compared With Midazolam) Study Group: Dexmedetomidine vs midazolam for sedation of critically ill patients: A randomized trial. JAMA 2009; 301:489 499 20. Senoglu N, Oksuz H, Dogan Z, et al: Sedation during noninvasive mechanical ventilation with dexmedetomidine or midazolam: A randomized, double-blind, prospective study. Curr Ther Res Clin Exp 2010; 71:141 153 21. Longrois D, Conti G, Mantz J, et al: Sedation in non-invasive ventilation: Do we know what to do (and why)? Multidiscip Respir Med 2014; 9:56 22. Hsu YW, Cortinez LI, Robertson KM, et al: Dexmedetomidine pharmacodynamics: Part I: Crossover comparison of the respiratory effects of dexmedetomidine and remifentanil in healthy volunteers. Anesthesiology 2004; 101:1066 1076 23. Morel DR, Forster A, Gemperle M: Noninvasive evaluation of breathing pattern and thoraco-abdominal motion following the infusion of ketamine or droperidol in humans. Anesthesiology 1986; 65:392 398 24. Miller AC, Jamin CT, Elamin EM: Continuous intravenous infusion of ketamine for maintenance sedation. Minerva Anestesiol 2011; 77:812 820 25. Honey BL, Harrison DL, Gormley AK, et al: Evaluation of adverse events noted in children receiving continuous infusions of dexmedetomidine in the intensive care unit. J Pediatr Pharmacol Ther 2010; 15:30 37 26. Lardieri AB, Fusco NM, Simone S, et al: Effects of clonidine on withdrawal from long-term dexmedetomidine in the pediatric patient. J Pediatr Pharmacol Ther 2015; 20:45 53 Pediatric Critical Care Medicine www.pccmjournal.org 7