Successful implementation of a pediatric sedation protocol for mechanically ventilated patients*

Similar documents
Interaction between Sedation and Weaning: How to Balance Them? Guillermo Castorena MD Fundacion Clinica Medica Sur Mexico

From the Department of Pharmacy (JM, CAF) and Department of Pulmonary and Critical

STARSHIP WITHDRAWAL OF ANALGESIA AND SEDATION

Ventilator-Associated Event Prevention: Innovations

KICU Spontaneous Awakening Trial (SAT) Questionnaire

Collaborative Regional Benchmarking Group (North of England, North Yorkshire & Humber and West Yorkshire)

Disclosure. Hospira Pharmaceuticals. Unrestricted research funding Honoraria for CME education administered via France Foundation

PICU Sedation Holidays. Jorge G. Sainz MD FAAP Medical Director PICU Medical Director Transport

Critical Care Strategic Clinical Network Provincial ICU Delirium Framework

Randomized controlled trial of interrupted versus continuous sedative infusions in ventilated children

Improving the Management of Pain, Agitation, and Delirium (PAD) in the Intensive Care Unit: Translating Evidence Into Practice

Goals for sedation during mechanical ventilation

Randomized controlled trial of daily interruption of sedatives in critically ill children

Sedation and delirium- drugs and clinical management

DELIRIUM IN ICU: Prevention and Management. Milind Baldi

POST-INTUBATION ANALGESIA AND SEDATION. August 2012 J Pelletier

Ventilated children are often sedated in order

Can Goal Directed Sedation Improve Outcomes?

The provision of adequate sedation and analgesia is a crucial

Sedation and Analgesia in the Critically Ill

Managing Delirium: The best way to achieve clarity (of mind) Tim Walsh. Professor of Critical Care, Edinburgh University

Canadian Practices for the Treatment of Delirium. Lisa Burry, BScPharm, PharmD

Implementing the WAT-1: an 11 item withdrawal assessment tool

Prolonged opioid therapy in the Critically Ill Pediatric Patient

ICU Liberation for the Pharmacist. A. Kendall Gross, PharmD, BCPS, BCCCP Critical Care Pharmacist UCSF Medical Center

Sedation Hold/Interruption and Weaning Protocol ( Wake-up and Breathe )

PHYSICIAN'S ORDERS Mark in for desired orders. If is blank, order is inactive. VENTILATOR SEDATION / ANALGESIC / DELIRIUM ORDER

Protocolized Sedation vs Usual Care in Pediatric Patients Mechanically Ventilated for Acute Respiratory Failure. Supplement 2 Table of Contents

Early Goal Directed Sedation In Critically Ill Patients

Sedation of the Critically Ill Patient

Pain & Sedation Management in PICU. Marut Chantra, M.D.

ICU Delirium in Infants & Children: Cause for Concern or False Alarm. Objectives

Dexmedetomidine: the various roles and utilization strategies. Julie Belfer, PharmD September 2014

Sleep Apnea and ifficulty in Extubation. Jean Louis BOURGAIN May 15, 2016

Fighting the Fog A Collaborative Approach to Decreasing ICU Delirium

Kendiss Olafson MD FRCPC MPH Section of Critical Care University of Manitoba

9/28/2016. Sedation Strategies in the ICU. Outline. ICU sedation. Recent clinical practice guidelines Top 10 myths A practical approach

Liberation from Mechanical Ventilation in Critically Ill Adults

Optimal sedation and management of anxiety in patients undergoing endobronchial ultrasound (EBUS)

Supplementary appendix

FOR REPRESENTATIVE EDUCATION

Sarah V. Cogle, PharmD, BCCCP Assistant Clinical Professor Auburn University Harrison School of Pharmacy Auburn, AL ALSHP Annual Clinical Meeting

ADMINISTRATIVE POLICY AND PROCEDURE MANUAL. Subject: Moderate Sedation/Analgesia- Procedural ( Conscious Sedation ) Policy

NURSING DEPARTMENT CRITICAL CARE POLICY MANUAL CRITICAL CARE PROTOCOL USE OF PROPOFOL (DIPRIVAN) FOR VENTILATOR MANAGEMENT

DEEP SEDATION TEST QUESTIONS

Optimize vent weaning and SBT outcomes. Identify underlying causes for SBT failures. Role SBT and weaning protocol have in respiratory care

SEDATION / ANALGESIA for Brain Failure Patient INASNACC

Sedation is a dynamic process.

New approaches of sedation in critically ill patients.

Afundamental tenet of caring for the critically CURRENT ISSUES IN AGITATION MANAGEMENT * PROCEEDINGS. I. Larry Cohen, MD, FCCP, FCCM ABSTRACT

INTERRUPTION OF SEDATIVE INFUSIONS IN CRITICALLY ILL PATIENTS UNDERGOING MECHANICAL VENTILATION

MORPHINE ADMINISTRATION

Critical Care Pharmacological Management of Delirium

Pain Management in the NICU. Tamorah Lewis MD, PhD

ARTICLE IN PRESS. doi: /j.jemermed TRAUMA PATIENTS CAN SAFELY BE EXTUBATED IN THE EMERGENCY DEPARTMENT

Do benzos, opioids, or strong anticholinergics cause delirium? Lisa Burry

Interprofessional Trauma Conference September 28th 2018 Montreal

The Efficacy of the COMFORT Scale in Assessing Optimal Sedation in Critically Ill Children Requiring Mechanical Ventilation

Dr Alireza Yarahmadi and Dr Arvind Perathur Mercy Medical Center - Winter Retreat Des Moines February 2012

Multiple Choice Questions

Sedation in Children

Clarification of Drug Allergy Information Using a Standardized Drug Allergy Questionnaire and Interview

Interfacility Protocol Protocol Title:

CUSP 4 MVP-VAP Patient Care Bundle

Subspecialty Rotation: Anesthesia

Analgesic-Sedatives Drug Dose Onset

Jennifer Mando-Vandrick, PharmD, BCPS Clinical Pharmacist, Emergency Department Director, PGY2 Critical Care Pharmacy Residency Duke University

Appendix 1. University of Minnesota Amplatz Children s Hospital Opioid Weaning Guideline

Results of a one-year, retrospective medication use evaluation. Joseph Ladd, PharmD PGY-1 Pharmacy Resident BHSF Homestead Hospital

Effectiveness and Safety of the Awakening and Breathing Coordination, Delirium

Kingdom; 2 University of Cambridge, Cambridge, United Kingdom

Surviving Sepsis Campaign. Guidelines for Management of Severe Sepsis/Septic Shock. An Overview

Complicated Withdrawal

Analgesia and Sedation Guidelines for Paediatric Intensive Care Unit

Difficult weaning from mechanical ventilation

Sedation and Delirium Questions

Conducting Delirium Research

+ Change in baseline mental status, inattention, and either disorganized thinking or altered level of consciousness. Delirium. Disclosure.

ROTATION SUMMARY PEDIATRIC PAIN MANAGEMENT

SEDATION FOR SMALL PROCEDURES

Withdrawal of Care in the ICU

Pain Module. Opioid-RelatedRespiratory Depression (ORRD)

A protocol of no sedation for critically ill patients receiving mechanical ventilation: a randomised trial

Pain Management at Stony Brook Medicine

Weaning and extubation in PICU An evidence-based approach

I. Subject. Moderate Sedation

Comparison of Procedural Sedation for the Reduction of Dislocated Total Hip Arthroplasty

Pain Assessment. Cathy Murray MSN RN OCNS-C Clinical Nurse Specialist December /21/2014 1

Digital RIC. Rhode Island College. Linda M. Green Rhode Island College

Implementation of a Delirium Screening Tool in a Pediatric Intensive Care Unit

WEANING READINESS & SPONTANEOUS BREATHING TRIAL MONITORING

Lumbar Fusion. Reference Guide for PACU CLINICAL PATHWAY. All patient variances to the pathway are to be circled and addressed in the progress notes.

Weaning: The key questions

VANDERBILT UNIVERSITY MEDICAL CENTER MULTIDISCIPLINARY SURGICAL CRITICAL CARE TRAUMA INTENSIVE CARE UNIT GLYCEMIC CONTROL PROTOCOL

Supplementary Online Content

Date 8/95; Rev.12/97; 7/98; 2/99; 5/01, 3/03, 9/03; 5/04; 8/05; 3/07; 10/08; 10/09; 10/10 Manual of Administrative Policy Source Sedation Committee

CRITICALLY APPRAISED PAPER (CAP)

The bispectral index (BIS) monitor was developed

(1996) 2002 JAMA. IDM

Transcription:

Successful implementation of a pediatric sedation protocol for mechanically ventilated patients* Kristina H. Deeter, MD; Mary A. King, MD, MPH; Debra Ridling, CCRN; Gretchen L. Irby, PharmD; Anne M. Lynn, MD; Jerry J. Zimmerman, MD, PhD, FCCM Objective: To evaluate the effect of a nursing-driven sedation protocol for mechanically ventilated pediatric patients on duration of use of analgesic and sedative medications. We hypothesized that a protocol would decrease length of sedation use and decrease days of mechanical ventilation and length of stay. Design: Retrospective cohort study with historical controls. Setting: Thirty-one-bed tertiary care, medical-surgical-cardiac pediatric intensive care unit in a metropolitan university-affiliated children s hospital. Patients: Children requiring mechanical ventilation longer than 48 hrs not meeting exclusion criteria. Interventions: Before protocol implementation, sedation was managed per individual physician orders. During the intervention period, analgesia and sedation were managed by nurses following an algorithm-based sedation protocol based on a comfort score. Measurements and Main Results: The observation group included consecutive patients admitted during the 12-month period before protocol education and implementation (n 153). The intervention group included patients admitted during the 12 months following protocol implementation (n 166). The median duration of total sedation days (intravenous plus enteral) was 7 days for the observation period and 5 days for the intervention period (p.026). Specifically, the median duration of morphine infusion was 6 days for the observation period and 5 days for the intervention period (p.015), whereas the median duration of lorazepam infusion was 2 days for the observation period and 0 days for the intervention period. After adjusting for severity of illness with the pediatric risk of mortality III (PRISM III) score, the Cox proportional hazards regression analysis demonstrated that at any point in time, patients in the intervention group were 23% more likely to be off all sedation (heart rate 0.77, p.020). Additionally, the intervention group tended to be associated with fewer days of mechanical ventilation (heart rate 0.81, p.060) and decreased pediatric intensive care unit length of stay (heart rate 0.81, p.058), although these associations did not quite reach statistical significance. Conclusion: A pediatric sedation protocol can significantly decrease days of benzodiazepine and opiate administration, which may improve pediatric intensive care unit resource utilization. (Crit Care Med 2011; 39:683 688) KEY WORDS: sedation protocol; pediatric sedation; continuous process improvement; morphine; lorazepam; opiates; benzodiazepines One of the most common and important interventions in providing care for critically ill children is the alleviation of pain and anxiety. The selection and method of administering sedative and analgesic medications to accomplish this goal is highly variable. Historically it has been based on individual physician preference and subjective nursing assessment of pain and anxiety. Infusions of analgesic and sedative agents are frequently used in intensive *See also p. 887. From the Departments of Pediatrics (KHD, MAK, DR, GLI, AML, JJZ) and Anesthesiology (AML), Seattle Children s Hospital University of Washington, Seattle, WA. This study was supported, in part, by the National Institutes of Health. The authors have not disclosed any potential conflicts of interest. For information regarding this article, E-mail: kdeeter@fcpeds.com Copyright 2011 by the Society of Critical Care Medicine and Lippincott Williams & Wilkins DOI: 10.1097/CCM.0b013e318206cebf care units (ICUs) as a means to provide a continuous level of comfort to critically ill patients (1, 2). This approach has been proven to decrease the discomfort associated with mechanical ventilation (MV), traumatic and surgical wounds, invasive devices, and procedures. In addition, analgesics and sedatives decrease oxygen consumption, modulate the intensity of the stress response, foster patient safety in a potentially dangerous ICU environment (by reducing risks of agitation-related injury and dislodgement of critical invasive devices), and facilitate bedside nursing care (2 5). Despite these benefits, continuous infusions of analgesics and sedatives have been identified as independent predictors of longer duration of MV as well as extended ICU and hospital length of stay (LOS) (6, 7). Prolonged sedation has been associated with increased procedures, acquired neuromuscular disorders, delirium, and posttraumatic stress disorder (8). Lastly, the long-term consequences of analgesics and sedatives on the developing brains of children are incompletely understood. Of particular concern, anesthetic drugs that alter synaptic transmission at -aminobutyrate type A and/or N-methyl-D-aspartate glutamate receptors cause neuroapoptosis in the developing brain and subsequent neurocognitive impairment in neonatal animal models (9 11). There is growing evidence that a sedation protocol for mechanically ventilated patients may decrease morbidity, LOS, and time on MV for critical care patients. Although there are published guidelines and many protocol examples for adult patients in the literature (8, 12 14), there is a paucity of practical evidence for children (15, 16). Adapting published adult protocols for pediatric patients is challenging, and the safety and efficacy of a sedation and weaning protocol in children is unknown, although it is currently being studied in a multi-institutional randomized, controlled trial (The Randomized Evaluation of Sedation Titration for Respiratory Failure [RESTORE] study, NCT00814099; http://clinicaltrials.gov). With a strategy Crit Care Med 2011 Vol. 39, No. 4 683

supported by continuous process improvement principles and Lean methodology in use at our institution (17), our multidisciplinary research team sought to design a pediatric sedation protocol that would be safe and effective and could be rapidly implemented in our large pediatric ICU (PICU). Our hypothesis was that a nursing-driven pediatric sedation protocol would decrease exposure to analgesics and sedatives, decrease ICU LOS, and decrease days of MV. METHODS Hour 1: After admission and/or intubation: Hour 2: Every 4 hours: 0400 0800 1200 1600 2000 2400 Every 12 hours: 0400 1600 After 24 hours of ventilation: Prior to extubation: Protocol Development. A multidisciplinary team that included PICU physicians, nurses, and a pharmacist met regularly over a period of 6 months to develop the Seattle Children s Comfort Protocol (SCCP) (Fig. 1). The team extensively reviewed published literature, order sets and protocols available from other institutions, and recommendations from individual experts in the field, such as Seattle Children s Hospital Pain Service physicians. Consensus was reached by all members of the team to continue with accepted institution preference for morphine and lorazepam due to staff comfort, experience, and clinical success with these medications. We did not reach team consensus on inclusion of a daily interruption of sedation in the initial protocol, which is often found in adult protocols (18, 19), and as such it was not included. Concerns included timing of the interruption, variability of awakening time, ability to provide 1:1 nursing to closely watch each patient during awakening, safety and risk of self-extubation, and variable pediatric developmental level as related to ability to understand verbal commands and calming. It was agreed that one protocol based on a comfort score would be developed in contrast to many adult protocols that separate pain and anxiety. To maximize compliance and facilitate education, we decided to continue with use of our institution s comfort scoring system, the Seattle PICU Comfort Score (Fig. 2), although there are other nationally recognized and validated comfort scores available for pediatrics (20, 21). A specific goal comfort score is ordered on morning interdisciplinary rounds depending on the depth of sedation preferred for an individual patient. For most patients, a score of 2 is typically appropriate, although as they near extubation, this order may be adjusted to a 3. Once the protocol was finalized, all staff completed 1 hr of required small group training that included a pharmacology review and direction on the use of the order set and SCCP. During the first week of implementation, a nurse educator was assigned to work individually with each bedside nurse to answer questions and ensure the SCCP was being properly implemented. Pharmacy staff took responsibility for daily auditing of protocol adherence, order accuracy, and reviewing compliance with titration instructions. Study Design and Data Collection. Early in the protocol development process, the team Bolus morphine 0.1 mg/kg (MAX 4mg per dose) and start infusion at 20 mcg/kg/hr. If already on infusion from OR or ER, continue infusion at current rate. Bolus morphine 0.1 mg/kg IV q15 minutes during first hour (MAX 4mg per dose) to goal comfort score. If 3 boluses are needed, then increase drip by 5 mcg/kg/hr. May give lorazepam 0.1mg/kg once (MAX 2mg/dose) prn anxiety. YES Continue current infusion rate. Bolus morphine 0.1 mg/kg q15 min prn to goal comfort score (MAX 4mg). YES Continue infusion with boluses as needed: -- Morphine 0.1 mg/kg q1hr prn pain -- Lorazepam 0.1 mg/kg q2 hr prn anxiety YES Decrease drip by 5 mcg/kg/hr. Continue boluses prn. If on lorazepam infusion, decrease this first may also decrease both at the same time. Achieving comfort goals? Reassess every 4 hrs. Were < 3 boluses required in a 4 hour time period? Reassess at 0400 and 1600. Maintaining goal comfort score? If > 4 lorazepam boluses in 12 hr period, may notify MD to order lorazepam drip to start at 10 mcg/kg/hr. Increase by 5 mcg/kg/hr immediately after 3rd bolus given in 6 hour period. Decrease by 5 mcg/kg/hr at 0400 and 1600 if no boluses given. Consider increasing goal comfort score. Ask MD to order PICU Extubation Orderset AND PICU Opioid and Benzodiazepine Weaning Orderset. Ask MD to decrease morphine and lorazepam prn doses by half and space to q2 and q4 prn respectively. NO May give lorazepam 0.1 mg/kg once (MAX 2mg call MD for order). Continue morphine boluses q15 min. If > 3 boluses given, increase infusion rate by 5 mcg/kg/hr. NO Increase infusion by 5mcg/kg/hr immediately after third bolus required in each 4 hour period. Continue morphine 0.1mg/kg q1 prn pain. Notify MD if at max 100 mcg/kg/hr Use lorazepam 0.1 mg/kg q2 hr prn anxiety. NO Discuss with MD. Consider increasing max dose or using alternative medications (Fentanyl, Dilaudid, Dexmedetomidine). Continue increasing drip every time 3 boluses are required in 4 hour time period. Figure 1. Seattle Children s Comfort Protocol. PICU, pediatric intensive care unit; MD, medical doctor; prn, as needed; IV, intravenous; OR, operating room; ER, emergency room; PCA, patient-controlled analgesia; q, every. 0 Patient receiving neuromuscular blockage 1 Deep sleep, heavily sedated, minimal movement, or unconscious 2 Sleepy, calm, arousable, cooperative with care or age appropriate 3 Tearing, restless, startles easily, fussy, but consolable 4 Guarding, anxious, may be grimacing, uncooperative with care, does not calm easily 5 Agitated, crying, tense posture or guarding, grimacing, not accepting mechanical ventilation (infants may exhibit apnea with pain) 6 Dangerously agitated, pulling at tubes and lines, requires restraints Figure 2. Seattle Pediatric Intensive Unit (PICU) Comfort Score. decided to objectively review the impact of the sedation protocol on the clinical care of our patient population. This research was conducted at Seattle Children s Hospital, a university-affiliated metropolitan children s hos- 684 Crit Care Med 2011 Vol. 39, No. 4

pital, and approved by the Institutional Review Board. Primary outcome measure was days of exposure to sedative and analgesic medications. This included benzodiazepines and opiates administered in intravenous (IV) form to maintain comfort and ventilator synchrony as well as enteral opiates (morphine and methadone) and benzodiazepines (typically lorazepam) dosed on a scheduled basis to prevent or treat symptoms of withdrawal. Secondary outcome measures were duration of MV and PICU LOS. Inclusion criteria: Admission to the ICU (medical, surgical, and cardiac); age newborn to 21 yrs; and need for MV for more than 2 days. Exclusion criteria: Diagnoses of seizures, acute or chronic neurologic dysfunction; extracorporeal life support; tracheostomy in place for any part of the ICU admission; infusion of a neuromuscular blocking agent; transfer from another ICU; or death during admission. Each subject met all inclusion and no exclusion criteria. During the observation period, patients were managed by using independent physician-directed approaches requiring specific physician s orders for sedation selection, dosing, and titration. During the intervention period (following healthcare staff education and training), children admitted to the ICU already intubated or who required intubation during admission were placed on the SCCP, although only those patients requiring MV for longer than 2 days were included in our retrospective review. Medical record numbers for patients meeting inclusion criteria were obtained from the virtual PICU system (vpicu, http://www.vpicu.org) for both the observation period (January 1 to December 31, 2007) and intervention period (May 1, 2008 to April 30, 2009). Data abstracted from electronic charts included inclusion and exclusion criteria, age, sex, diagnosis, Pediatric Risk of Mortality III (PRISM III) score (22), LOS, duration of MV, IV sedation including medication and duration, enteral sedation including medication and duration, and whether or not the patient had undergone surgery. Total days of sedation (reported in tables as IV plus enteral) included the IV infusions received during hospitalization in addition to any oral benzodiazepines or opiates given on a scheduled basis while admitted or prescribed at discharge. Outpatient medications were tracked with our electronic records system for refills, and outpatient physician notes were reviewed to determine exact length of time required for weaning medications (typically oral morphine, methadone, or lorazepam). There were only a small number of patients who required outpatient weaning, and all of these records were available for review to confirm date of cessation of weaning medication (most followed by the cardiac, transplant, or oncology services). During the intervention period, we also reviewed charts for physician orders for the SCCP and noted any reasons for protocol noncompliance. Statistical Analysis. Baseline patient characteristics of the two study groups were compared by using descriptive statistics. We observed a non-normal distribution of the study patient outcome measures (total sedation days, duration of MV, and LOS) when the data were plotted as histograms. Outcome measures were reported as both mean and median values with respective standard deviations and interquartile ranges. Wilcoxon s rank sum test was used to compare continuous nonparametric outcome measures between groups. Similarly, given the non-normal distribution of the data, a Cox proportional hazards regression analysis was performed to assess the differences between the intervention group and the traditional group after adjustment for pediatric severity of illness (using a PRISM III score). The hazard ratio represents the likelihood at any point in time that a child in the intervention group (as compared with the observation group) would be on a given therapy. All statistical tests were two-sided. Data were analyzed by using STATA 10 (Stata, College Station, TX). RESULTS Observation period ICU admits = 1388 Intervention period ICU admits = 1074 MV > 2 days = 285 MV > 2 days = 325 Patients. Two hundred eighty-five patients met inclusion criteria during the observation period. After exclusions were made, 153 patient charts were analyzed (Fig. 3). The records of 325 consecutive patients meeting inclusion criteria during the intervention period were reviewed, and data from 166 patients were ultimately analyzed after review for exclusion criteria. The demographics of both groups were similar (Table 1) as were reasons for study exclusion. There were 30 patients in the intervention group that would have met criteria to be placed on the SCCP but were excluded by the ordering physician due to preference 46 6 15 8 17 40 N/A Acute/chronic neurologic disease Transfer from another ICU Tracheostomy in place PCA/epidural Required ECLS (survived) Expired during admission ** MD exclusion 153 TOTAL PATIENTS INCLUDED 166 Figure 3. Subject flow chart with exclusions. **Includes patients meeting other exclusion criteria, i.e., on extracorporeal life support before death. ICU, intensive care unit; MV, mechanical ventilation; PCA, patient-controlled analgesia; ECLS, extracorporeal life support. 30 11 28 11 10 39 30 for alternative narcotic, concerns for profound hypotension or heart failure, reintubation or readmission before previous narcotic weans, parent refusal, or morphine allergy. Median PRISM III score for these patients was comparable with study populations at 6.5. Of note, more than half of all exclusions took place in the first 4 months of protocol introduction with the remainder in the following 8 months as staff became more comfortable with use of the SCCP. Outcomes. The duration of sedation exposure, duration of MV, and PICU LOS were compared by study group (Table 2). Patients in the intervention group were exposed to significantly fewer days of morphine infusion, lorazepam infusion, and total sedation (IV plus enteral) than patients in the observation group. For the primary outcome, the intervention group (as compared with the observation group) was 23% more likely to be off all sedation (heart rate 0.77, 95% confidence interval: 0.61 0.96, p.020). For secondary analyses, the intervention group tended to be associated with fewer days of MV (heart rate 0.81, p.060) and PICU LOS (heart rate 0.81, p.058), although these associations did not quite reach statistical significance (Table 3). The rate of unplanned extubations during each study period was also retrospectively reviewed and summarized. In 2007, during the observation period, there were 13 documented unplanned extubations out of 4,003 ventilator days (an average of 0.32 events per 100 ventilator days). During the first year of protocol implementation, there were 11 unplanned extubations out of 4,737 ventilator days (average of 0.23 events per 100 ventilator days). The national benchmark Crit Care Med 2011 Vol. 39, No. 4 685

Table 1. Comparison of baseline characteristics by intervention group Demographic Observation Group (n 153) (http://mmp-bench.com) during these time periods was 0.65 accidental extubations per 100 days. These data include all ventilated patients admitted to our ICU, including those patients meeting criteria for this study. Lastly, we collected data on dexmedetomidine use during our study period. Dexmedetomidine was used in 74 of 153 patients (48%) in the observation group and only 40 of 166 patients (25%) in the intervention group during some point in the course of their ICU therapy. We found a strong association between dexmedetomidine use (yes/no) and total sedation days (median) in the observation group (dexmedetomidine yes: 12 days vs. dexmedetomidine no: 6 days, p.018). We found no association between dexmedetomidine use and total sedation days in the intervention group (dexmedetomidine yes: 5 days vs. dexmedetomidine no: 5 days, p.244). DISCUSSION Intervention Group (n 166) Age (yrs) Mean (SD) 3.0 (5.0) 2.6 (4.7) Median (interquartile range) 0.5 (0.1 3.1) 0.4 (0.1 1.7) Gender Male 85 (56%) 88 (53%) Female 68 (44%) 78 (47%) Diagnostic category Surgical (%) 105 (69%) 108 (65%) Medical (%) 48 (31%) 58 (35%) Pediatric risk of mortality III score Mean (SD) 6.9 (5.5) 6.2 (4.8) Median (interquartile range) 6 (3 10) 5 (3 9) Table 2. Comparison of days of pediatric intensive care unit therapy by intervention group Variable (Days) Observation Group (n 153) Intervention Group (n 166) p a Morphine infusion Mean (SD) 9.9 (11.0) 7.0 (6.3) Median (IQR) 6 (4 12) 5 (3 9).015 Lorazepam infusion Mean (SD) 6.6 (11.2) 1.6 (3.7) Median (IQR) 2 (0 7) 0 (0 2).001 Total sedation (intravenous enteral) b Mean (SD) 15.9 (19.6) 11.6 (12.6) Median (IQR) 7 (4 22) 5 (3 17).026 Mechanical ventilation Mean (SD) 8.1 (9.0) 6.3 (5.4) Median (IQR) 5 (3 9) 5 (3 7).155 Pediatric intensive care unit length of stay Mean (SD) 13.1 (13.3) 10.4 (7.7) Median (IQR) 9.5 (5.4 15.2) 8.2 (5 13.2).302 IQR, interquartile range. a p values are calculated via Wilcoxon s rank sum test; b total sedation days are defined as total days of sedative medications, including all routes (intravenous and enteral) as well as all locations (in the intensive care unit, the pediatric ward, and home). Table 3. Cox proportional hazards regression model to assess the effect of intervention group on duration of therapy, after adjustment for severity of illness Variable (Days) Hazard Ratio (95% Confidence Interval) p Total sedation (intravenous enteral) 0.77 (0.61 0.96).020 Mechanical ventilation 0.81 (0.65 1.01).060 Pediatric intensive care unit length of stay 0.81 (0.64 1.01).058 Implementation of a nursing-driven pediatric sedation protocol significantly decreased the duration of both opiate and benzodiazepine continuous infusions as well as total duration of sedative exposure. As found in previous adult studies, a multidisciplinary team approach was successfully used to create a sedation protocol that empowers intensive care nurses to safely administer and titrate continuous infusions of opiates and benzodiazepines for critically ill children. When we accounted for the potential confounding effect of severity of illness as measured by PRISM III score, our data suggest that a pediatric sedation protocol very likely can also decrease duration of MV and ICU LOS. Importantly, we found no increase in the rate of unplanned extubations. We found that dexmedetomidine use fell by approximately 50% in our study population, suggesting that children required fewer alternative sedation therapies while on the SCCP; however, this would need future study to verify. We also found no association between dexmedetomidine use and total sedation days (our primary outcome measure) in patients on the SCCP. The positive changes seen in our unit are likely a result of many factors: Implementation of a nursing-driven protocol, education of the healthcare team with respect to the medications used for pain and sedation, and a growing awareness of the importance of reducing unnecessary sedation. This is a single center study in a teaching hospital; our protocol may not be easily translatable to other PICUs. The SCCP was designed to meet the needs of our unit and was created with input from our nursing staff and from specialists utilizing our services. Similar centers will likely need to construct individual protocols to meet their own needs. We encountered many challenges in developing our protocol. In reviewing published protocols, we found that many centers use different medication algorithms for pain and anxiety. As discrimination of pain and anxiety in children is difficult, we instead decided to focus on a single comfort protocol to address both variables with titration guided by a comfort scale. The Seattle PICU Comfort Tool was developed at our institution, has been in use in the ICU for approximately 10 yrs, and thus was well known to staff. Although this tool is not a nationally validated scoring system, it was derived from the adult Ramsay sedation tool (23) and developed as part of a quality improvement project with inter-rater reliability testing completed before use. An- 686 Crit Care Med 2011 Vol. 39, No. 4

other challenge in developing the SCCP was to standardize sedation provided to both our medical and surgical patients with a single algorithm in an effort to decrease risk of error given that nurses may work in both the medical/surgical and cardiac ICUs. After much collaborative discussion, we chose to utilize morphine as the primary sedation agent for all patients on the protocol. The Seattle Children s team of anesthesiologists, intensivists, and pharmacists collaboratively adopted the use of morphine years ago due to its analgesic and sedative properties, its pediatric safety and efficacy profile, relatively slow development of tachyphylaxis, and its cost-effectiveness. Common concerns regarding histamine release with morphine has been shown to be less problematic with infusions (24, 25). Use of morphine and lorazepam for longterm sedation has also been supported by recent consensus guidelines (8). Particularly when dosing is high, weaning of long-term sedation can require days to months, and this may prolong duration of MV and hospitalization (26). Children may be sent home on oral analgesic/ sedative medications and weaning protocols that create risk for patient and family safety concerns. A key component of the SCCP is a rapid attainment of comfort with bolus dosing and titration of the infusion, but with subsequent mandatory assessment every 4 hrs and reduction of the infusion recommended every 12 hrs once the patient is comfortable. In this way, patients are slowly, but steadily, weaned from sedation before extubation. Aside from minimizing sedation duration, we have also demonstrated how a pediatric ICU sedation protocol can decrease days of MV and ICU LOS, although our data in these regards did not quite reach statistical significance given the small nature of this single-center study. This decrease in utilization of scarce pediatric medical resources may impart a large cost savings to the patient, the hospital, and society. Further prospective study with large numbers of subjects utilizing pediatric tools for assessment of delirium and posttraumatic stress disorder along with long-term follow-up of neurodevelopmental effects is needed to fully answer such questions. From a hospital perspective, we hope to improve safe patient flow through the ICU, decrease costs related to sedation, decrease costs related to complications in the ICU or ward related to drug exposure, and improve staff satisfaction through use of standardization and guidelines (27, 28). Patient deaths during each time period were excluded in this study to further describe a change in the total days of sedation, MV, and LOS. Median PRISM III score for patients who expired in the observation group was 16.5 and for the intervention group was 11. Many of these patients required extracorporeal life support or a paralytic infusion thereby removing them from protocol inclusion. Many others were found to have fatal malignancies, failed bone marrow or solid organ transplants, or inoperable cardiac conditions. Deaths were reviewed by authors and circumstances found to be similar between groups. Although it is impossible to know without further review and alternative study design, there was no documentation discovered in these charts implicating the method of sedation as a contributing factor to death. Limitations We cannot generalize our results to the critically ill child with neurologic disease or the child who is so ill as to require extracorporeal life support or go on to die as those populations were excluded from our study. Specifically, another agent such as fentanyl may be preferable for patients who have severe hemodynamic compromise or who require frequent neurologic exams, and we did not test the efficacy of morphine in these populations. Our retrospective design and use of historical controls is another important limitation to our study as practice and patient populations may change over time. A retrospective design was deemed necessary as the research team felt that attempting to train only half of our entire healthcare team in an effort to randomize to different arms of a study would not be feasible in our unit and would lead to contamination of results during the intervention phase. Unfortunately, our retrospective electronic chart review was also not adequate to capture total milligrams of sedatives and narcotics administered daily. It is possible that even though the total days of sedative administration decreased, the actual amount (in milligrams) of these medications may have increased as has been reported by other investigators using similar protocols (19). Total amount of drug exposure (in milligrams), incidence of withdrawal symptoms and delirium (29), and number of children requiring weaning regimens following discharge from the ICU should be studied in a prospective fashion. CONCLUSION The development and implementation of a pediatric comfort protocol that addresses analgesia and sedation is feasible and safe. Initiation of this quality improvement protocol significantly decreased exposure of critically ill children to analgesic and sedative medications with an associated reduction in days of MV and ICU LOS. The key components of this protocol include the use of a consistent comfort metric, nursing education and empowerment, and forced, scheduled assessments with defined responses. ACKNOWLEDGMENTS The authors would like to thank Tellen Bennett, Lin DiGennaro, Lauraine Heer, and John Salyer for their assistance with technical aspects of this study and manuscript preparation. REFERENCES 1. Jenkins I: The provision of analgesia and sedation in the PICU: Current practice and recent advances. Paediatr Anaest 2002; 12: 837 839 2. Jacobs JR, Reves JG, Glass PS: Rationale and technique for continuous infusions in anesthesia. Int Anesthesiol Clin 1991; 29:23 38 3. Buck ML, Blumer JL: Opioids and other analgesics. Adverse effects in the intensive care unit. Crit Care Clin 1991; 7:615 637 4. Mehta S, Burry L, Fischer S, et al: Canadian survey of the use of sedatives, analgesics, and neuromuscular blocking agents in critically ill patients. Crit Care Med 2006; 34:374 380 5. Anand KJ, Ingraham J: Pediatric. Tolerance, dependence, and strategies for compassionate withdrawal of analgesics and anxiolytics in the pediatric ICU. Crit Care Nurse 1996; 16:87 93 6. Kollef MH, Levy NT, Ahrens TS, et al: The use of continuous i.v. sedation is associated with prolongation of mechanical ventilation. Chest 1998; 114:541 548 7. 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 2132 8. Jacobi J, Fraser GL, Coursin DB, et al: Clinical practice guidelines for the sustained use of sedatives and analgesics in the critically ill adult. Crit Care Med 2002; 30:119 141 9. Loepke AW: Developmental neurotoxicity of sedatives and anesthetics: A concern for neonatal and pediatric critical care medicine? Pediatr Crit Care Med 2010; 11:217 226 10. Fredriksson A, Pontén E, Gordh T, et al: Neonatal exposure to a combination of N-methyl- Crit Care Med 2011 Vol. 39, No. 4 687

D-aspartate and gamma-aminobutyric acid type A receptor anesthetic agents potentiates apoptotic neurodegeneration and persistent behavioral deficits. Anesthesiology 2007; 107: 427 436 11. Creeley CE, Olney JW: The young: Neuroapoptosis induced by anesthetics and what to do about it. Anesth Analg 2010; 110:442 448 12. Brook AD, Ahrens TS, Schaiff R, et al: Effect of a nursing-implemented sedation protocol on the duration of mechanical ventilation. Crit Care Med 1999; 27:2609 2615 13. Kress JP, Pohlman AS, Hall JB: Sedation and analgesia in the intensive care unit. Am J Respir Crit Care Med 2002; 166:1024 1028 14. Girard TD, Kress JP, Fuchs BD, et al: Efficacy and safety of a paired sedation and ventilator weaning protocol for mechanically ventilated patients in intensive care (Awakening and Breathing Controlled Trial): a randomised controlled trial. Lancet 2008; 371:126 134 15. Playfor S, Jenkins I, Boyles C, et al: Consensus guidelines on sedation and analgesia in critically ill children. Intensive Care Med 2006; 32:1125 1136 16. Hartman ME, McCrory DC, Schulman SR: Efficacy of sedation regimens to facilitate mechanical ventilation in the pediatric intensive care unit: A systematic review. Pediatr Crit Care Med 2009; 10:246 255 17. Black J: The Toyota Way to Healthcare Excellence. Chicago, IL, Health Administration Press, 2008 18. Kress JP, Pohlman AS, O Connor MF, Hall JB: Daily interruption of sedative infusions in critically ill patients undergoing mechanical ventilation. N Engl J Med 2000; 342:1471 1477 19. de Wit M, Gennings C, Jenvey WI, et al: Randomized trial comparing daily interruption of sedation and nursing-implemented sedation algorithm in medical intensive care unit patients. Crit Care Med 2008; 12:R70 20. 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 21. Ambuel B, Hamlett KW, Marx CM, Blumer JL: Assessing distress in pediatric intensive care environments: the COMFORT scale. J Pediatr Psychol 1992; 17:95 109 22. Pollack MM, Patel KM, Ruttimann UE: PRISM III: An updated Pediatric Risk of Mortality score. Crit Care Med 1996; 24:743 752 23. Ramsay MA, Savege TM, Simpson BR, et al: Controlled sedation with alphaxalonealphadolone. BMJ 1974; 2:656 659 24. Warner MA, Hosking MP, Gray JR, et al: Narcotic-induced histamine release: A comparison of morphine, oxymorphone, and fentanyl infusions. J Cardiothorac Vasc Anesth 1991; 5:481 484 25. Doenicke A, Moss J, Lorenz W, et al: Intravenous morphine and nalbuphine increase histamine and catecholamine release without accompanying hemodynamic changes. Clin Pharmacol Ther 1995; 58:81 89 26. Randolph AG, Wypij D, Venkataraman ST, et al: Effect of mechanical ventilator weaning protocols on respiratory outcomes in infants and children: A randomized controlled trial. JAMA 2002; 288:2561 2568 27. Arias-Rivera S, Sánchez-Sánchez Mdel M, Santos-Díaz R, et al: Effect of a nursingimplemented sedation protocol on weaning outcome. Crit Care Med 2008; 36:2054 2060 28. Clemmer TP, Spuhler VJ: Developing and gaining acceptance for patient care protocols. New Horiz 1998; 6:12 19 29. Smith HA, Fuchs DC, Pandharipande PP, et al: Delirium: An emerging frontier in the management of critically ill children. Crit Care Clin 2009; 25:593 614, x 688 Crit Care Med 2011 Vol. 39, No. 4